WO2018140923A1 - Methods of treating cancer - Google Patents

Methods of treating cancer Download PDF

Info

Publication number
WO2018140923A1
WO2018140923A1 PCT/US2018/015874 US2018015874W WO2018140923A1 WO 2018140923 A1 WO2018140923 A1 WO 2018140923A1 US 2018015874 W US2018015874 W US 2018015874W WO 2018140923 A1 WO2018140923 A1 WO 2018140923A1
Authority
WO
WIPO (PCT)
Prior art keywords
tumor
pharmaceutical composition
fenoterol
cancer
expression
Prior art date
Application number
PCT/US2018/015874
Other languages
French (fr)
Inventor
Irving W. Wainer
Original Assignee
Mitchell Woods Pharmaceuticals, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Mitchell Woods Pharmaceuticals, Inc. filed Critical Mitchell Woods Pharmaceuticals, Inc.
Publication of WO2018140923A1 publication Critical patent/WO2018140923A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • A61K31/135Amines having aromatic rings, e.g. ketamine, nortriptyline
    • A61K31/137Arylalkylamines, e.g. amphetamine, epinephrine, salbutamol, ephedrine or methadone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca

Definitions

  • the present disclosure relates to methods of treating cancer by reducing tumor generated L-lactate in the microenvironment surrounding a tumor by administration of at least one agent, such as for example a fenoterol analogue.
  • Cancer is the second leading cause of human death next to coronary disease in the
  • fenoterol analogues are used to improve the effectiveness of one or more immune checkpoint blockade therapy by reducing tumor generated L-lactate in the microenvironment surrounding a tumor.
  • the exemplary methods described herein can be used, for example, to improve the effectiveness of an immune checkpoint blockade therapy in the treatment of pancreatic cancer, breast cancer, or other cancers.
  • the method includes administering a therapeutically effective amount of a fenoterol analogue to a cancer patient to improve the effectiveness of one or more immune checkpoint blockade therapies.
  • the fenoterol analogue is an antagonist of pyruvate kinase M2 (PKM2). In embodiments, the fenoterol analogue is an antagonist of hexokinase-2 (HK2). In embodiments, the fenoterol analogue is a compound that attenuates monocarboxylate transporter 4 (MCT4) expression and/or function, thereby decreasing L-lactate export and increasing L-lactate concentrations within the tumor.
  • PLM2 pyruvate kinase M2
  • HK2 hexokinase-2
  • MCT4 monocarboxylate transporter 4
  • fenoterol analogues include one or more compounds selected from the group consisting of (R,R')-4'-methoxy- 1-naphthylfenoterol ("MNF"), (R,S')-4'- methoxy- 1-naphthylfenoterol, (R,R')-ethylMNF, (R,R')-napthylfenoterol, (R,S napthylfenoterol, (R,R')-ethyl-naphthylfenoterol, (R,R')-4' -amino- 1-naphthylfenoterol, (R,R')- 4'-hydroxy-l-naphthylfenoterol, (R,R')-4-methoxy-ethylfenoterol, (R,R')-methoxyfenoterol, (R,R')- ethylf
  • the fenoterol analogue is (R,R')-4'-methoxy- 1-naphthylfenoterol (MNF), a compound having the formula:
  • the presently described methods include administering a therapeutically effective amount of a pharmaceutical composition containing a fenoterol analogue and a pharmaceutically acceptable carrier to a cancer patient to decrease the amount of L-lactate in a tumor microenvironment.
  • the cancer patient is known to have pancreatic or breast cancer.
  • the method includes administering one or more therapeutic agents in addition to a fenoterol analogue.
  • the methods can include administration of the one or more therapeutic agents separately, sequentially or concurrently, for example in a combined composition with a fenoterol analogue.
  • the one or more therapeutic agents administered in addition to a fenoterol analogue may be one or more immune checkpoint blockade therapy.
  • a method of attenuating monocarboxylate transporter 4 (MCT4) expression and/or function is described. Attenuating MCT4 expression and/or function decreases L-lactate export out of cancer cells and increases L-lactate concentrations in cancer cells.
  • the method includes administering a therapeutically effective amount of a fenoterol analogue to attenuate MCT4 expression and/or function.
  • a method of decreasing the amount (and hence activity) of hexokinase-2 (HK2) in cancer cells showing increased HK2 expression is described. Decreasing the amount HK2 decreases glucose metabolism and glycolysis and increases the sensitivity of cancer cells to cell death inducers.
  • the method includes administering a therapeutically effective amount of a fenoterol analogue to decrease expression of HK2.
  • a method of decreasing the amount (and hence activity) of glucose transporter facilitators e.g., Glut-1, Glut-8, and others
  • the method includes administering a therapeutically effective amount of a fenoterol analogue to decrease expression of one or more glucose transporter facilitators.
  • Fig. 1 shows that (R,S')-MNF reduces plasma L-lactate concentrations in mice bearing PANC-1 tumor xenografts
  • Fig. 2 shows that (R,S')-MNF dose-dependently reduces ⁇ -catenin expression in
  • Fig. 3 shows that (R,R')-MNF inhibits glucose uptake and lactate production in
  • FIG. 4 shows that (R,R')-MNF reduces lactate production in the breast cancer cell lines MCF-7 and MDA-MB-231;
  • Figure 5 shows the pathway of aerobic glycolysis in cancer cells resulting in an enhanced uptake of glucose and production of L-lactate ("the Warburg effect");
  • FIG. 6 shows that (R,R')-MNF reduces the expression of HK2 and the L-lactate transporter MCT4 in tumor tissue obtained from the PANC1 xenograft;
  • Figure 7 shows that (R,R')-MNF reduces the expression of ⁇ -catenin in tumor tissues obtained from mice bearing a PANC-1 xenograft;
  • Figure 8 shows that in mice with a PANC-1 xenograft tumor, (R,R')-MNF decreases the plasma concentration of L-lactate and increases the tumor L-lactate concentration;
  • Figure 9 shows the impact of lactate on tumor microenvironment. .
  • Cancer cells evolve several alterations in their metabolism to survive in unfavorable microenvironments, while retaining their ability to proliferate.
  • metabolic reprogramming is a key aspect of tumorigenesis and has a profound effect on gene expression, cellular differentiation, metastasis, and tumor microenvironment.
  • One metabolic adaptation of tumor cells is a shift to aerobic glycolysis as a main source of ATP, rather than oxidative phosphorylation (OXPHOS), irrespective of oxygen availability, a phenomenon referred to as the Warburg effect.
  • This phenotype may promote a state of apoptosis resistance, the generation of biosynthetic precursors for proliferation, and increased invasive ability.
  • aerobic glycolysis is a major component of metabolic reprograming and is characterized by enhanced glucose uptake and its conversion to L-lactate via the glycolytic pathway.
  • PKM2 pyruvate kinase M2
  • HK2 glycolytic enzyme hexokinase 2
  • Warburg effect in cancer cells showing increased HK2 expression. Depletion of HK2 has been shown to restore oxidative glucose metabolism and increase sensitivity to cell death inducers such as radiation and chemotherapies.
  • Another key aspect of metabolic reprogramming is the MCT4-mediated export of
  • L-lactate In pancreatic cancer, increased expression of MCT4 is associated with a poor prognosis due to the role that increased L-lactate concentration plays in immunoresistance. Tumor-generated L-lactate inhibits anticancer immune response through decreased cytotoxic activity of T lymphocytes and natural killer cells.
  • pancreatic cancer cells e.g., PANC-1 cells
  • a fenoterol analog in accordance with the methods of the present disclosure attenuates glycolysis by reducing the expression of Glut-1, hexokinase II, PKM2, and lactate dehydrogenase.
  • Administration of a fenoterol analog in accordance with the methods of the present disclosure reduces L-lactate plasma concentration relative to pre-dose concentrations. Since immunotherapy lacks therapeutic efficacy in pancreatic cancer and some of the observed resistance has been attributed to the tumor microenvironment, reduction of L-lactate concentration in the tumor microenvironment in accordance with the presently described methods potentiates the effect of checkpoint inhibitors and therapeutic vaccines.
  • a PKM2 antagonist such as a fenoterol analogue
  • a disease state by reducing tumor generated L-lactate in the environment surrounding a tumor.
  • This change in the microenvironment surrounding the tumor may reduce cellular proliferation, the expression of proteins key to the survival of cancer cells, and the resistance of cancer cells to treatment by anticancer drugs (known as multidrug resistance).
  • a compound that decreases HK2 expression and activity such as a fenoterol analogue, is used to treat a disease state.
  • cancer is treated by restoring oxidative glucose metabolism and increasing sensitivity of cancer cells to cell death inducers.
  • a compound that attenuates MCT4 expression and/or function such as a fenoterol analogue, is used to treat a disease state.
  • the MCT4-mediated export of L-lactate is reduced, thereby reducing tumor-generated L-lactate in the environment surrounding a tumor enhancing anticancer immune response and increasing cytotoxic activity of T lymphocytes and natural killer cells.
  • the compounds described herein can be used to treat pancreatic or breast cancer as well as other forms of cancer. Based upon these findings, methods of treating disorders and diseases modulated by glycolysis are described.
  • AM251 l-(2,4-dichlorophenyl)-5-(4-iodophenyl)-4-methyl-N-(l-piperidyl)pyrazole- 3- carboxamide
  • AM630 l-[2-(morpholin-4-yl)ethyl]-2-methyl-3-(4-methoxybenzoyl)-6-iodoindole
  • AR adrenergic receptor
  • EGFR epidermal growth factor receptor
  • ERK extracellular regulated kinase
  • GPR55 G protein-coupled receptor 55
  • GPCR G protein-coupled receptor
  • ICI 118,551 3-(isopropylamino)-l-[(7-methyl-4-indanyl)oxy]butan-2-ol
  • IP intraperitoneal
  • IV intravenous
  • UV ultraviolet
  • Acyl A group of the formula RC(O)- wherein R is an organic group.
  • Acyloxy A group having the structure -OC(0)R, where R may be an optionally substituted alkyl or optionally substituted aryl. "Lower acyloxy” groups are those where R contains from 1 to 10 (such as from 1 to 6) carbon atoms.
  • Administration To provide or give a subject a composition, such as a pharmaceutical composition including one or more fenoterol analogues by any effective route. Exemplary routes of administration include, but are not limited to, injection (such as subcutaneous, intramuscular, intradermal, intraperitoneal (“IP”), and intravenous (“IV”)), oral, sublingual, rectal, transdermal, intranasal, vaginal and inhalation routes.
  • injection such as subcutaneous, intramuscular, intradermal, intraperitoneal (“IP”), and intravenous (“IV”)
  • oral sublingual, rectal, transdermal, intranasal, vaginal and inhalation routes.
  • Alkoxy A radical (or substituent) having the structure -O-R, where R is a substituted or unsubstituted alkyl.
  • Methoxy (-OCH 3 ) is an exemplary alkoxy group.
  • R is alkyl substituted with a non-interfering substituent.
  • Thioalkoxy refers to -S-R, where R is substituted or unsubstituted alkyl.
  • Haloalkyloxy means a radical -OR where R is a haloalkyl.
  • Alkoxy carbonyl A group of the formula -C(0)OR, where R may be an optionally substituted alkyl or optionally substituted aryl.
  • “Lower alkoxy carbonyl” groups are those where R contains from 1 to 10 (such as from 1 to 6) carbon atoms.
  • Alkyl An acyclic, saturated, branched- or straight-chain hydrocarbon radical, which, unless expressly stated otherwise, contains from one to fifteen carbon atoms; for example, from one to ten, from one to six, or from one to four carbon atoms. This term includes, for example, groups such as methyl, ethyl, n-propyl, isopropyl, isobutyl, t-butyl, pentyl, heptyl, octyl, nonyl, decyl, or dodecyl.
  • the term "lower alkyl” refers to an alkyl group containing from one to ten carbon atoms.
  • alkyl groups can either be unsubstituted or substituted.
  • An alkyl group can be substituted with one or more substituents (for example, up to two substituents for each methylene carbon in an alkyl chain).
  • alkyl substituents include, for instance, amino groups, amide, sulfonamide, halogen, cyano, carboxy, hydroxy, mercapto, trifluorom ethyl, alkyl, alkoxy (such as methoxy), alkylthio, thioalkoxy, arylalkyl, heteroaryl, alkylamino, dialkylamino, alkylsulfano, keto, or other functionality.
  • Amino carbonyl (carbamoyl): A group of the formula C(0)N(R)R', wherein R and R are independently of each other hydrogen or a lower alkyl group.
  • p2-adrenergic receptor A subtype of adrenergic receptors that are members of the G-protein coupled receptor family. P2-AR subtype is involved in respiratory diseases, cardiovascular diseases, premature labor and, as disclosed herein, tumor development. Increased expression of p2-ARs can serve as therapeutic targets.
  • Cannabinoid Receptors A class of cell membrane receptors under the G protein- coupled receptor superfamily. The cannabinoid receptors contain seven transmembrane spanning domains. Cannabinoid receptors are activated by three major groups of ligands, endocannabinoids (produced by the mammalian body), plant cannabinoids (such as THC, produced by the cannabis plant) and synthetic cannabinoids (such as HU-210). All of the endocannabinoids and plant cannabinoids are lipophilic, i.e., fat soluble, compounds. Two subtypes of cannabinoid receptors are CBi (see GenBank Accession No.
  • the CB 2 receptor is expressed mainly in the immune system and in hematopoietic cells. Additional non-CBi and non-CB 2 include GPR55 (GenBank Accession No. NM_005683.3 or NP_005674.2 protein, each of which is hereby incorporated by reference as of May 23, 2012), GPR119 (GenBank Accession No.
  • NM_178471.2 or NP_848566.1 protein each of which is hereby incorporated by reference as of May 23, 2012
  • GPR18 also known as N- arachidonyl glycine receptor and involved in microglial migration, GenBank Accession No. NM_001098200 mRNA, NP_001091670.1, each of which is hereby incorporated by reference as of May 23, 2012).
  • CBi and CB 2 receptors are about 44% similar. When only the transmembrane regions of the receptors are considered, amino acid similarity between the two receptor subtypes is approximately 68%. In addition, minor variations in each receptor have been identified. Cannabinoids bind reversibly and stereo-selectively to the cannabinoid receptors. The affinity of an individual cannabinoid to each receptor determines the effect of that cannabinoid. Cannabinoids that bind more selectively to certain receptors are more desirable for medical usage.
  • GPR55 is coupled to the G-protein G i3 and/or Gn and activation of the receptor leads to stimulation of rhoA, cdc42 and rack GPR55 is activated by the plant cannabinoids A 9 - THC and cannabidiol, and the endocannabinoids anandamide, 2-AG, noladin ether in the low nanomolar range.
  • CBi and CB 2 receptors are coupled to inhibitory G proteins. This indicates that both types of receptors will have different readouts. For example, activation of CBi causes apoptosis whereas increase in GPR55 activity is oncogenic.
  • the CBi receptor antagonist (also termed 'inverse agonist') compound, AM251 is, in fact, an agonist for GPR55. It binds GPR55 and is readily internalized. This illustrates the opposite behavior of these two GPCRs.
  • Carbamate A group of the formula -OC(0)N(R)-, wherein R is H, or an aliphatic group, such as a lower alkyl group or an aralkyl group.
  • Chemotherapy; chemotherapeutic agents any chemical agent with therapeutic usefulness in the treatment of diseases characterized by abnormal cell growth. Such diseases include tumors, neoplasms, and cancer as well as diseases characterized by hyperplastic growth.
  • a chemotherapeutic agent is an agent of use in treating neoplasms such as solid tumors, including a tumor associated with CB receptor activity and/or expression.
  • a chemotherapeutic agent is radioactive molecule.
  • a CB receptor regulator such as one or more fenoterol analogues or a combination thereof is a chemotherapeutic agent.
  • a chemotherapeutic agent is carmustine, lomustine, procarbazine, streptozocin, or a combination thereof.
  • a chemotherapeutic agent of use e.g., see Slapak and Kufe, Principles of Cancer Therapy, Chapter 86 in Harrison's Principles of Internal Medicine, 14th edition; Perry et al., Chemotherapy, Ch. 17 in Abel off, Clinical Oncology 2 nd ed., ⁇ 2000 Churchill Livingstone, Inc; Baltzer L., Berkery R.
  • control or Reference Value A "control” refers to a sample or standard used for comparison with a test sample.
  • the control is a sample obtained from a healthy subject or a tissue sample obtained from a patient diagnosed with a disorder or disease, such as a tumor, that did not respond to treatment with a p2-agonist.
  • the control is a historical control or standard reference value or range of values.
  • Derivative A chemical substance that differs from another chemical substance by one or more functional groups. In embodiments, a derivative retains a biological activity of a molecule from which it was derived.
  • Effective amount An amount of agent that is sufficient to generate a desired response, such as reducing or inhibiting one or more signs or symptoms associated with a condition or disease. When administered to a subject, a dosage will generally be used that will achieve target tissue concentrations. In some examples, an "effective amount" is one that treats one or more symptoms and/or underlying causes of any of a disorder or disease.
  • an "effective amount” is a "therapeutically effective amount” in which the agent alone with an additional therapeutic agent(s) (for example a chemotherapeutic agent) induces the desired response such as treatment of a tumor.
  • a desired response is to decrease tumor size or metastasis in a subject to whom the therapy is administered. Tumor metastasis does not need to be completely eliminated for the composition to be effective.
  • a composition can decrease metastasis by a desired amount, for example by at least 20%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, at least 98%, or even at least 100%) (elimination of the tumor), as compared to metastasis in the absence of the composition.
  • a composition can decrease the number of cancer cells by a desired amount, for example by at least 20%, at least 50%, at least 60%>, at least 70%, at least 80%), at least 90%, at least 95%, at least 98%, or even at least 100% (elimination of detectable cancer cells), as compared to the number of cancer cells in the absence of the composition.
  • the effective amount of a composition useful for reducing, inhibiting, and/or treating a disorder in a subject will be dependent on the subject being treated, the severity of the disorder, and the manner of administration of the therapeutic composition.
  • Effective amounts a therapeutic agent can be determined in many different ways, such as assaying for a reduction in tumor size or improvement of physiological condition of a subject having a tumor, such as a brain tumor. Effective amounts also can be determined through various in vitro, in vivo or in situ assays.
  • Fenoterol analogues include (R,R')-4'-methoxy-l- naphthylfenoterol ("MNF"), (R,S')-4'-methoxy-l-naphthylfenoterol, (R,R')-ethylMNF, (R,R')- napthylfenoterol, (R,S')-napthylfenoterol, (R,R')-ethyl-naphthylfenoterol, (R,R' )-4' -amino- 1- naphthylfenoterol, (R,R')-4'-hydroxy-l-naphthylfenoterol, (R,R')-4'-methoxy-ethylfenoterol, (R,R')-methoxyfenoterol, (R,R')-ethylfenoterol, (R,R'')-ethylfenote
  • Inflammation When damage to tissue occurs, the body's response to the damage is usually inflammation.
  • the damage may be due to trauma, lack of blood supply, hemorrhage, autoimmune attack, transplanted exogenous tissue or infection.
  • This generalized response by the body includes the release of many components of the immune system (for instance, IL-1 and TNF), attraction of cells to the site of the damage, swelling of tissue due to the release of fluid and other processes.
  • Isomers Compounds that have the same molecular formula but differ in the nature or sequence of bonding of their atoms or the arrangement of their atoms in space are termed “isomers”. Isomers that differ in the arrangement of their atoms in space are termed “stereoisomers”. Stereoisomers that contain two or more chiral centers and are not mirror images of one another are termed “diastereomers.” Steroisomers that are non-superimposable mirror images of each other are termed "enantiomers.” When a compound has an asymmetric center, for example, if a carbon atom is bonded to four different groups, a pair of enantiomers is possible.
  • An enantiomer can be characterized by the absolute configuration of its asymmetric center and is described by the R- and S-sequencing rules of Cahn and Prelog, or by the manner in which the molecule rotates the plane of polarized light and designated as dextrorotatory or levorotatory (i.e., as (+) or (-) isomers, respectively).
  • a chiral compound can exist as either an individual enantiomer or as a mixture thereof. A mixture containing equal proportions of the enantiomers is called a "racemic mixture.”
  • the compounds described herein may possess one or more asymmetric centers; such compounds can therefore be produced as individual (R), (S), (R,R), (R,S'), (S,R') and (S,S')-stereoisomers or as mixtures thereof.
  • R the description or naming of a particular compound in the specification and claims is intended to include both individual enantiomers and mixtures, racemic or otherwise, thereof.
  • the methods for the determination of stereochemistry and the separation of stereoisomers are well known in the art (see, e.g., March, Advanced Organic Chemistry, 4th edition, New York: John Wiley and Sons, 1992, Chapter 4).
  • compositions and formulations suitable for pharmaceutical delivery of one or more therapeutic compounds or molecules such as one or more nucleic acid molecules, proteins or antibodies that bind these proteins, and additional pharmaceutical agents.
  • parenteral formulations usually comprise injectable fluids that include pharmaceutically and physiologically acceptable fluids such as water, physiological saline, balanced salt solutions, aqueous dextrose, glycerol or the like as a vehicle.
  • pharmaceutically and physiologically acceptable fluids such as water, physiological saline, balanced salt solutions, aqueous dextrose, glycerol or the like as a vehicle.
  • physiologically acceptable fluids such as water, physiological saline, balanced salt solutions, aqueous dextrose, glycerol or the like
  • solid compositions for example, powder, pill, tablet, or capsule forms
  • conventional non-toxic solid carriers can include, for example, pharmaceutical grades of mannitol, lactose, starch, or magnesium stearate.
  • compositions to be administered can contain minor amounts of non-toxic auxiliary substances, such as wetting or emulsifying agents, preservatives, and pH buffering agents and the like, for example sodium acetate or sorbitan monolaurate.
  • non-toxic auxiliary substances such as wetting or emulsifying agents, preservatives, and pH buffering agents and the like, for example sodium acetate or sorbitan monolaurate.
  • Phenyl groups may be unsubstituted or substituted with one, two or three substituents, with substituent(s) independently selected from alkyl, heteroalkyl, aliphatic, heteroaliphatic, thioalkoxy, halo, haloalkyl (such as -CF 3 ), nitro, cyano, -OR (where R is hydrogen or alkyl), -N(R)R (where R and R are independently of each other hydrogen or alkyl), -COOR (where R is hydrogen or alkyl) or -C(0)N(R)R" (where R and R" are independently selected from hydrogen or alkyl).
  • purified does not require absolute purity; rather, it is intended as a relative term.
  • a purified preparation is one in which a desired component such as an (R,R ')-enantiomer of fenoterol is more enriched than it was in a preceding environment such as in a (+)-fenoterol mixture.
  • a desired component such as (R,R')-enantiomer of fenoterol is considered to be purified, for example, when at least about 70%, 80%, 85%, 90%, 92%), 95%), 97%), 98%), or 99% of a sample by weight is composed of the desired component.
  • Purity of a compound may be determined, for example, by high performance liquid chromatography (HPLC) or other conventional methods.
  • HPLC high performance liquid chromatography
  • the fenoterol analogue enantiomers are purified to represent greater than 90%, often greater than 95% of the other enantiomers present in a purified preparation.
  • the purified preparation may be essentially homogeneous, wherein other stereoisomers are less than 1%.
  • a compound described herein may be obtained in a purified form or purified by any of the means known in the art, including silica gel and/or alumina chromatography. See, e.g., Introduction to Modern Liquid Chromatography, 2nd Edition, ed. by Snyder and Kirkland, New York: John Wiley and Sons, 1979; and Thin Layer Chromatography, ed. by Stahl, New York: Springer Verlag, 1969.
  • a compound includes purified fenoterol or fenoterol analogue with a purity of at least about 70%, 80%, 85%, 90%, 92%, 95%, 97%, 98%, or 99% of a sample by weight relative to other contaminants.
  • a compound includes at least two purified stereoisomers each with a purity of at least about 70%, 80%, 85%, 90%, 92%, 95%), 97%), 98%), or 99% of a sample by weight relative to other contaminants.
  • a compound can include a substantially purified (R,R')-fenoterol analogue and a substantially purified (R,S')-fenoterol analogue.
  • Subject includes both human and veterinary subjects, for example, humans, non-human primates, dogs, cats, horses, rats, mice, and cows.
  • mammal includes both human and non-human mammals.
  • Tissue A plurality of functionally related cells.
  • a tissue can be a suspension, a semi-solid, or solid.
  • Tissue includes cells collected from a subject such as the brain or a portion thereof.
  • Tumor All neoplastic cell growth and proliferation, whether malignant or benign, and all pre-cancerous and cancerous cells and tissues.
  • a primary tumor is tumor growing at the anatomical site where tumor progression began and proceeded to yield this mass.
  • under conditions sufficient for includes administering one or more fenoterol analogues to a subject to at a concentration sufficient to allow the desired activity.
  • the desired activity is reducing or inhibiting a sign or symptom associated with a disorder or disease, such as a breast or pancreatic, can be evidenced, for example, by a delayed onset of clinical symptoms of the tumor in a susceptible subject, a reduction in severity of some or all clinical symptoms of the tumor, a slower progression of the tumor (for example by prolonging the life of a subject having the tumor), a reduction in the number of tumor reoccurrence, an improvement in the overall health or well-being of the subject, or by other parameters well known in the art that are specific to the particular disease.
  • the desired activity is preventing or inhibiting tumor growth, such as breast cancer or pancreatic cancer growth.
  • Tumor growth does not need to be completely inhibited for the treatment to be considered effective.
  • a partial reduction or slowing of growth such as at least about a 10% reduction, such as at least 20%, at least about 30%, at least about 40%, at least about 50% or greater is considered to be effective.
  • Fenoterol analogues useful in the methods herein include (R,R')-4'-methoxy-l- naphthylfenoterol ("MNF"), (R,S')-4'-methoxy-l-naphthylfenoterol, (R,R')-ethylMNF, (R,R')- napthylfenoterol, (R,S')-napthylfenoterol, (R,R')-ethyl-naphthylfenoterol, (R,R' )-4' -amino- 1- naphthylfenoterol, (R,R')-4'-hydroxy-l-naphthylfenoterol, (R,R')-4'-methoxy-ethylfenoterol, (R,R')-4'-methoxyfenoterol, (R,R')-ethylfenoterol, (R,R,R,
  • Examples of suitable groups for R1-R3 that can be cleaved in vivo to provide a hydroxy group include, without limitation, acyl, acyloxy and alkoxy carbonyl groups.
  • Compounds having such cleavable groups are referred to as "prodrugs.”
  • the term "prodrug,” as used herein, means a compound that includes a substituent that is convertible in vivo (e.g., by hydrolysis) to a hydroxyl group.
  • Various forms of prodrugs are known in the art, for example, as discussed in Bundgaard, (ed.), Design of Prodrugs, Elsevier (1985); Widder, et al. (ed.), Methods in Enzymology, Vol.
  • administering comprises administering a therapeutically effective amount of MNF, NF or a combination thereof. In some embodiments, administering comprises administering a therapeutically effective amount of MNF.
  • Solvate means a physical association of a compound with one or more solvent molecules. This physical association involves varying degrees of ionic and covalent bonding, including by way of example covalent adducts and hydrogen bonded solvates. In certain instances the solvate will be capable of isolation, for example when one or more solvent molecules are incorporated in the crystal lattice of the crystalline solid. "Solvate” encompasses both solution-phase and isolable solvates. Representative solvates include ethanol- associated compound, methanol-associated compounds, and the like. "Hydrate” is a solvate wherein the solvent molecule(s) is/are H 2 0.
  • the disclosed compounds also encompass salts including, if several salt-forming groups are present, mixed salts and/or internal salts.
  • the salts are generally pharmaceutically acceptable salts that are non-toxic. Salts may be of any type (both organic and inorganic), such as fumarates, hydrobromides, hydrochlorides, sulfates and phosphates. In an example, salts include non-metals (e.g., halogens) that form group VII in the periodic table of elements. For example, compounds may be provided as a hydrobromide salt.
  • Additional examples of salt-forming groups include, but are not limited to, a carboxyl group, a phosphonic acid group or a boronic acid group, that can form salts with suitable bases.
  • salts can include, for example, nontoxic metal cations, which are derived from metals of groups IA, IB, IIA and IIB of the periodic table of the elements.
  • alkali metal cations such as lithium, sodium or potassium ions, or alkaline earth metal cations such as magnesium or calcium ions can be used.
  • the salt can also be a zinc or an ammonium cation.
  • the salt can also be formed with suitable organic amines, such as unsubstituted or hydroxyl -substituted mono-, di- or tri-alkylamines, in particular mono-, di- or tri-alkylamines, or with quaternary ammonium compounds, for example with N-methyl-N- ethylamine, diethylamine, triethylamine, mono-, bis- or tris- (2-hydroxy-lower alkyl)amines, such as mono-, bis- or tris- (2- hydroxyethyl)amine, 2-hydroxy-tert-butylamine or tris(hydroxymethyl)methylamine, N,N-di-lower alkyl-N-(hydroxy-lower alkyl)amines, such as N,N-dimethyl-N-(2-hydroxyethyl)amine or tri-(2- hydroxyethyl)amine, or N-methyl-D- glucamine, or quaternary ammonium compounds such as te
  • Exemplary compounds disclosed herein possess at least one basic group that can form acid- base salts with inorganic acids.
  • basic groups include, but are not limited to, an amino group or imino group.
  • inorganic acids that can form salts with such basic groups include, but are not limited to, mineral acids such as hydrochloric acid, hydrobromic acid, sulfuric acid or phosphoric acid.
  • Basic groups also can form salts with organic carboxylic acids, sulfonic acids, sulfo acids or phospho acids or N-substituted sulfamic acid, for example acetic acid, propionic acid, glycolic acid, succinic acid, maleic acid, hydroxymaleic acid, methylmaleic acid, fumaric acid, malic acid, tartaric acid, gluconic acid, glucaric acid, glucuronic acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, salicylic acid, 4-aminosalicylic acid, 2- phenoxybenzoic acid, 2-acetoxybenzoic acid, embonic acid, nicotinic acid or isonicotinic acid, and, in addition, with amino acids, for example with a-amino acids, and also with methanesulfonic acid, ethanesulfonic acid, 2-hydroxymethanesulfonic acid, ethane- 1,2-disulfonic
  • employing a pharmaceutically acceptable salt may also serve to adjust the osmotic pressure of a composition.
  • the compounds used in the method are provided are polymorphous.
  • the compounds can be provided in two or more physical forms, such as different crystal forms, crystalline, liquid crystalline or non-crystalline (amorphous) forms.
  • any of the above described compounds e.g., (R,R') and/or (R,S') fenoterol analogues or a hydrate or pharmaceutically acceptable salt thereof
  • combinations thereof are intended for use in the manufacture of a medicament for treatment of breast or pancreatic cancer.
  • the disclosed fenoterol analogues can be synthesized by any method known in the art including those described in U.S. Patent Application Publication No. US 2010-0168245 Al, U.S. Patent Application Publication No. US 2012-0157543 Al and International Patent Publication No. WO 2011/112867, each of which is hereby incorporated by reference in its entirety.
  • Many general references providing commonly known chemical synthetic schemes and conditions useful for synthesizing the disclosed compounds are available (see, e.g., Smith and March, March's Advanced Organic Chemistry: Reactions, Mechanisms, and Structure, Fifth Edition, Wiley- Interscience, 2001; or Vogel, A Textbook of Practical Organic Chemistry, Including Qualitative Organic Analysis, Fourth Edition, New York: Longman, 1978).
  • Compounds as described herein may be purified by any of the means known in the art, including chromatographic means, such as UPLC (including chiral UPLC), preparative thin layer chromatography, flash column chromatography and ion exchange chromatography. Any suitable stationary phase can be used, including normal and reversed phases as well as ionic resins. Most typically the disclosed compounds are purified via open column chromatography or prep chromatography.
  • chromatographic means such as UPLC (including chiral UPLC), preparative thin layer chromatography, flash column chromatography and ion exchange chromatography.
  • Any suitable stationary phase can be used, including normal and reversed phases as well as ionic resins.
  • Most typically the disclosed compounds are purified via open column chromatography or prep chromatography.
  • Scheme I An exemplary synthesis of 4 stereoisomers of 1 - 6 including the coupling of the epoxide formed from either (R)- or (S)-3',5'-dibenzyloxyphenyl bromohydrin with the (R)- or (S)- enantiomer of the appropriate benzyl -protected 2-amino-3-benzylpropane (1 - 5) or the (R)- or (S)- enantiomer of N-benzyl-2-aminoheptane (6).
  • Scheme II Exemplary synthesis of (R)-7 and (S)-7 using 2-phenethylamine. resulting compounds may be deprotected by hydrogenation over Pd/C and purified as fumarate salts.
  • Scheme III describes an exemplary synthesis for the chiral building blocks used in
  • the disclosed fenoterol analogues can be useful, at least, for reducing or inhibiting one or more symptoms or signs associated with cancer. Accordingly, pharmaceutical compositions comprising at least one disclosed fenoterol analogue are also described herein.
  • Formulations for pharmaceutical compositions are well known in the art. For example, Remington's Pharmaceutical Sciences, by E. W. Martin, Mack Publishing Co., Easton, PA, 19th Edition, 1995, describes exemplary formulations (and components thereof) suitable for pharmaceutical delivery of (R,R')-fenoterol and disclosed fenoterol analogues. Pharmaceutical compositions comprising at least one of these compounds can be formulated for use in human or veterinary medicine. Particular formulations of a disclosed pharmaceutical composition may depend, for example, on the mode of administration (e.g., oral or parenteral) and/or on the disorder to be treated. In some embodiments, formulations include a pharmaceutically acceptable carrier in addition to at least one active ingredient, such as a fenoterol compound.
  • compositions useful for the disclosed methods and compositions are conventional in the art.
  • the nature of a pharmaceutical carrier will depend on the particular mode of administration being employed.
  • parenteral formulations usually comprise injectable fluids that include pharmaceutically and physiologically acceptable fluids such as water, physiological saline, balanced salt solutions, aqueous dextrose, glycerol or the like as a vehicle.
  • non-toxic solid carriers can include, for example, pharmaceutical grades of mannitol, lactose, starch, or magnesium stearate.
  • pharmaceutical compositions to be administered can optionally contain minor amounts of non-toxic auxiliary substances or excipients, such as wetting or emulsifying agents, preservatives, and pH buffering agents and the like; for example, sodium acetate or sorbitan monolaurate.
  • non-limiting excipients include, nonionic solubilizers, such as cremophor, or proteins, such as human serum albumin or plasma preparations.
  • compositions may be formulated as a pharmaceutically acceptable salt.
  • Pharmaceutically acceptable salts are non-toxic salts of a free base form of a compound that possesses the desired pharmacological activity of the free base. These salts may be derived from inorganic or organic acids. Non-limiting examples of suitable inorganic acids are hydrochloric acid, nitric acid, hydrobromic acid, sulfuric acid, hydriodic acid, and phosphoric acid.
  • Non-limiting examples of suitable organic acids are acetic acid, propionic acid, glycolic acid, lactic acid, pyruvic acid, malonic acid, succinic acid, malic acid, maleic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, methyl sulfonic acid, salicylic acid, formic acid, trichloroacetic acid, trifluoroacetic acid, gluconic acid, asparagic acid, aspartic acid, benzenesulfonic acid, p- toluenesulfonic acid, naphthalenesulfonic acid, and the like. Lists of other suitable pharmaceutically acceptable salts are found in Remington's Pharmaceutical Sciences, 19th Edition, Mack Publishing Company, Easton, PA, 1995. A pharmaceutically acceptable salt may also serve to adjust the
  • the dosage form of a disclosed pharmaceutical composition will be determined by the mode of administration chosen.
  • oral dosage forms may be employed.
  • Oral formulations may be liquid such as syrups, solutions or suspensions or solid such as powders, pills, tablets, or capsules. Methods of preparing such dosage forms are known, or will be apparent, to those skilled in the art.
  • compositions comprising a disclosed compound may be formulated in unit dosage form suitable for individual administration of precise dosages.
  • amount of active ingredient such as (R,R')-MNF or F administered will depend on the subject being treated, the severity of the disorder, and the manner of administration, and is known to those skilled in the art.
  • the formulation to be administered will contain a quantity of the extracts or compounds disclosed herein in an amount effective to achieve the desired effect in the subject being treated.
  • compositions are provided in the form of a tablet containing from about 1.0 to about 50 mg of the active ingredient, particularly about 2.0 mg, about 2.5 mg, 5 mg, about 10 mg, or about 50 mg of the active ingredient for the symptomatic adjustment of the dosage to the subject being treated.
  • a tablet containing from about 1 mg to about 50 mg (such as about 2 mg to about 10 mg) active ingredient is administered two to four times a day, such as two times, three times or four times.
  • a suitable dose for parental administration is about 1 milligram per kilogram (mg/kg) to about 100 mg/kg, such as a dose of about 10 mg/kg to about 80 mg/kg, such including about 1 mg/kg, about 2 mg/kg, about 5 mg/kg, about 20 mg/kg, about 30 mg/kg, about 40 mg/kg, about 50 mg/kg, about 80 mg/kg or about 100 mg/kg administered parenterally.
  • mg/kg milligram per kilogram
  • a dose of about 10 mg/kg to about 80 mg/kg such including about 1 mg/kg, about 2 mg/kg, about 5 mg/kg, about 20 mg/kg, about 30 mg/kg, about 40 mg/kg, about 50 mg/kg, about 80 mg/kg or about 100 mg/kg administered parenterally.
  • other higher or lower dosages also could be used, such as from about 0.001 mg/kg to about 1 g/kg, such as about 0.1 to about 500 mg/kg, including about 0.5 mg/kg to about 200 mg/kg.
  • compositions comprising one or more of the disclosed compositions can be carried out with dose levels and pattern being selected by the treating physician.
  • multiple doses are administered.
  • the composition is administered parenterally once per day.
  • the composition can be administered twice per day, three times per day, four times per day, six times per day, every other day, twice a week, weekly, or monthly. Treatment will typically continue for at least a month, more often for two or three months, sometimes for six months or a year, and may even continue indefinitely, i.e., chronically. Repeat courses of treatment are also possible.
  • the pharmaceutical composition is administered without concurrent administration of a second agent for the treatment of breast or pancreatic cancer.
  • one or more of the disclosed compositions is administered without concurrent administration of other agents, such as without concurrent administration of an additional agent also known to target the tumor.
  • a therapeutically effective amount of a disclosed pharmaceutical composition is administered concurrently with an additional agent, including an additional therapy.
  • the disclosed compounds are administered in combination with a chemotherapeutic agent, antioxidants, anti-inflammatory drugs or combinations thereof.
  • the disclosed compounds are administered in combination with an immune checkpoint therapy.
  • Immune checkpoints affect immune system functioning and can be stimulatory or inhibitory. Tumors can use these checkpoints to protect themselves from immune system attacks.
  • Checkpoint therapy can block inhibitory checkpoints, restoring immune system function.
  • the immune checkpoint therapy may impact the interaction between the transmembrane programmed cell death 1 protein (PDCDl, PD-1; also known as CD279) and its ligand, PD-1 ligand 1 (PD-L1, CD274). Cancer-mediated upregulation of PD-L1 on the cell surface may inhibit T cells that might otherwise attack.
  • PDCDl transmembrane programmed cell death 1 protein
  • PD-1 also known as CD279
  • PD-1 ligand 1 PD-1 ligand 1
  • Cancer-mediated upregulation of PD-L1 on the cell surface may inhibit T cells that might otherwise attack.
  • the presently disclosed fenoterol analogues may be administered with antibodies that bind to either PD-1 or PD-L1 and therefore block the interaction and thereby allow the T-cells to attack the tumor.
  • the presently disclosed fenoterol analogues may be administered with IgG4 PD1 antibody (such as, for example, antibody BGB-A317, Nivolumab or Pembrolizumab which may provide benefits with respect to one or more of melanoma, lung cancer, kidney cancer and Hodgkin's lymphoma).
  • the presently disclosed fenoterol analogues may be administered with a PD-L1 inhibitor, such as, for example, atezolizumab, avelumab, or and durvalumab.
  • a PD-L1 inhibitor such as, for example, atezolizumab, avelumab, or and durvalumab.
  • the presently disclosed fenoterol analogues may be administered with antibodies that block the immune checkpoint molecule CTLA-4, (such as, for example, ipilimumab, which may provide benefits with respect to lung cancer or pancreatic cancer, specifically in combination with other drugs).
  • CTLA-4 such as, for example, ipilimumab, which may provide benefits with respect to lung cancer or pancreatic cancer, specifically in combination with other drugs.
  • the presently disclosed fenoterol analogues may be administered with a combination of CTLA-4 blockade with PD-1 or PD-L1 inhibitors.
  • the presently disclosed fenoterol analogues may be administered with a therapeutic agent that targets an intrinsic checkpoint blockade, such as, for example, the gene encoding Cytokine-inducible SH2-containing protein (CISH).
  • a therapeutic agent that targets an intrinsic checkpoint blockade such as, for example, the gene encoding Cytokine-inducible SH2-containing protein (CISH).
  • a disclosed pharmaceutical composition is administered as adjuvant therapy.
  • a pharmaceutical composition containing one or more of the disclosed compounds is administered orally daily to a subject in order to prevent or retard tumor growth.
  • a composition containing equal portions of two or more disclosed compounds is provided to a subject.
  • a composition containing unequal portions of two or more disclosed compounds is provided to the subject.
  • a composition contains unequal portions of a (R,R')-fenoterol derivative and a (S,R')-fenoterol derivative and/or a (R,S') -derivative.
  • the composition includes a greater amount of the (S,R)- or (R,S')-fenoterol derivative.
  • Such therapy can be given to a subject for an indefinite period of time to inhibit, prevent, or reduce tumor reoccurrence.
  • the present disclosure includes methods of treating disorders including reducing or inhibiting one or more signs or symptoms associated with cancer, such as pancreatic cancer or breast cancer.
  • Presently disclosed methods include administering fenoterol, such as (R,R)- fenoterol, a disclosed fenoterol analogue or a combination thereof (and, optionally, one or more other pharmaceutical agents) depending upon the receptor population of the tumor, to a subject in a pharmaceutically acceptable carrier and in an amount effective to reduce tumor generated L- lactate in the environment surrounding cancer cells.
  • Treatment of a tumor includes preventing or reducing signs or symptoms associated with the presence of such tumor (for example, by reducing the size or volume of the tumor or a metastasis thereof).
  • Such reduced growth can in some examples decrease or slow metastasis of the tumor, or reduce the size or volume of the tumor by at least 10%, at least 20%, at least 50%, or at least 75%, such as between 10%-90%, 20%-80%, 30%- 70%, 40%-60%, including a 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 90%, or 95% reduction.
  • treatment includes reducing the invasive activity of the tumor in the subject, for example by reducing the ability of the tumor to metastasize.
  • treatment using the methods disclosed herein prolongs the time of survival of the subject.
  • Routes of administration useful in the disclosed methods include but are not limited to oral and parenteral routes, such as intravenous (IV), intraperitoneal (IP), rectal, topical, ophthalmic, nasal, and transdermal as described in detail above.
  • IV intravenous
  • IP intraperitoneal
  • rectal topical
  • ophthalmic nasal
  • transdermal transdermal
  • an effective amount of a disclosed fenoterol analogue will depend, at least, on the particular method of use, the subject being treated, the severity of the tumor, and the manner of administration of the therapeutic composition.
  • a "therapeutically effective amount" of a composition is a quantity of a specified compound sufficient to achieve a desired effect in a subject being treated. For example, this may be the amount of a fenoterol analogue necessary to prevent or inhibit tumor growth and/or one or more symptoms associated with the tumor in a subject.
  • a therapeutically effective amount of a disclosed fenoterol analogue is an amount sufficient to prevent or inhibit a tumor, such as a brain or liver tumor growth and/or one or more symptoms associated with the tumor in a subject without causing a substantial cytotoxic effect on host cells.
  • Therapeutically effective doses of a disclosed fenoterol compound or pharmaceutical composition can be determined by one of skill in the art, with a goal of achieving concentrations that are at least as high as the IC 50 of the applicable compound disclosed in the examples herein.
  • An example of a dosage range is from about 0.001 to about 10 mg/kg body weight orally in single or divided doses.
  • a dosage range is from about 0.005 to about 5 mg/kg body weight orally in single or divided doses (assuming an average body weight of approximately 70 kg; values adjusted accordingly for persons weighing more or less than average).
  • compositions are, for example, provided in the form of a tablet containing from about 1.0 to about 50 mg of the active ingredient, particularly about 2.5 mg, about 5 mg, about 10 mg, or about 50 mg of the active ingredient for the symptomatic adjustment of the dosage to the subject being treated.
  • a tablet containing from about 1 mg to about 50 mg active ingredient is administered two to four times a day, such as two times, three times or four times.
  • a suitable dose for parental administration is about 1 milligram per kilogram (mg/kg) to about 100 mg/kg, such as a dose of about 10 mg/kg to about 80 mg/kg, such including about 1 mg/kg, about 2 mg/kg, about 5 mg/kg, about 20 mg/kg, about 30 mg/kg, about 40 mg/kg, about 50 mg/kg, about 80 mg/kg or about 100 mg/kg administered parenterally.
  • mg/kg milligram per kilogram
  • a dose of about 10 mg/kg to about 80 mg/kg such including about 1 mg/kg, about 2 mg/kg, about 5 mg/kg, about 20 mg/kg, about 30 mg/kg, about 40 mg/kg, about 50 mg/kg, about 80 mg/kg or about 100 mg/kg administered parenterally.
  • other higher or lower dosages also could be used, such as from about 0.001 mg/kg to about 1 g/kg, such as about 0.1 to about 500 mg/kg, including about 0.5 mg/kg to about 200 mg/kg.
  • compositions comprising one or more of the disclosed compositions can be carried out with dose levels and pattern being selected by the treating physician.
  • multiple doses are administered.
  • the composition is administered parenterally once per day.
  • the composition can be administered twice per day, three times per day, four times per day, six times per day, every other day, twice a week, weekly, or monthly. Treatment will typically continue for at least a month, more often for two or three months, sometimes for six months or a year, and may even continue indefinitely, i.e., chronically. Repeat courses of treatment are also possible.
  • the specific dose level and frequency of dosage for any particular subject may be varied and will depend upon a variety of factors, including the activity of the specific compound, the metabolic stability and length of action of that compound, the age, body weight, general health, sex and diet of the subject, mode and time of administration, rate of excretion, drug combination, and severity of the condition of the subject undergoing therapy.
  • Subjects can be screened prior to initiating the disclosed therapies, for example to select a subject in need of or at risk of developing cancer.
  • the method can include screening subjects to determine if they have or are at risk of developing cancer, such as if the subject is in need of pancreatic cancer or breast cancer inhibition.
  • the cancer is regulated by at least one of B2-adrenergic receptor (AR) activity or expression, cannabinoid (CB) receptor activity or expression, or epidermal growth factor receptor (EGFR) activity or expression.
  • AR B2-adrenergic receptor
  • CB cannabinoid
  • EGFR epidermal growth factor receptor
  • Such cancers include, but are not limited to various types of breast cancer.
  • Subjects having a tumor that expresses B2-adrenergic receptor (AR), cannabinoid (CB) receptor (including but not limited to GPR55), and epidermal growth factor receptor (EGFR) or at risk of developing such a tumor are selected.
  • subjects are diagnosed with the tumor by clinical signs, laboratory tests, or both.
  • a subject in need of the disclosed therapies is selected by detecting a tumor expressing B2-adrenergic receptor (AR), cannabinoid (CB) receptor (including but not limited to GPR55), and epidermal growth factor receptor (EGFR) or regulated by their activity, such as by detecting B2-adrenergic receptor (AR) activity, cannabinoid (CB) receptor (including but not limited to GPR55) activity, and epidermal growth factor receptor (EGFR) activity or expression in a sample obtained from a subject identified as having, suspected of having or at risk of acquiring such a tumor.
  • AR B2-adrenergic receptor
  • CB cannabinoid receptor
  • EGFR epidermal growth factor receptor
  • detection of altered such as at least a 10% alteration, including a 10%-90%, 20%-80%, 30%-70%, 40%-60%, such as a 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 90%, 95% alteration or more in B2-adrenergic receptor (AR) expression or activity, cannabinoid (CB) receptor (including but not limited to GPR55) expression or activity, and epidermal growth factor receptor (EGFR) expression or activity as compared to B2-adrenergic receptor (AR) expression or activity, cannabinoid (CB) receptor (including but not limited to GPR55) expression or activity, and epidermal growth factor receptor (EGFR) expression or activity in the absence of a primary tumor, indicates that the tumor can be treated using the fenoterol compositions and methods provided herein.
  • Pre-screening is not required prior to administration of the therapeutic agents disclosed herein (such as those including fenoterol, a fenoterol analogue or a combination thereof).
  • subjects can be monitored for decreases in tumor growth, tumor volume or in one or more clinical symptoms associated with the tumor.
  • subjects are analyzed one or more times, starting 7 days following treatment.
  • Subjects can be monitored using any method known in the art including those described herein including imaging analysis. Additional Treatments and Additional Therapeutic Agents
  • a partial response is a reduction, such as at least a 10%, at least a 20%), at least a 30%>, at least a 40%, at least a 50%, or at least a 70% reduction in one or more signs or symptoms associated with the disorder or disease, or activity, including tumor size or volume.
  • the method further includes administering a therapeutic effective amount of a fenoterol analogue with additional therapeutic treatments.
  • the subject prior to, during, or following administration of a therapeutic amount of an agent that reduces tumor generated L-lactate in the environment surrounding the tumor, the subject can receive one or more other therapies.
  • the subject receives one or more treatments to remove or reduce the tumor prior to administration of a therapeutic amount of a composition including fenoterol, a fenoterol analogue or combination thereof.
  • Examples of such therapies include, but are not limited to, surgical treatment for removal or reduction of the tumor (such as surgical resection, cryotherapy, or chemoembolization), as well as anti-tumor pharmaceutical treatments which can include radiotherapeutic agents, anti-neoplastic chemotherapeutic agents, antibiotics, alkylating agents and antioxidants, kinase inhibitors, and other agents.
  • additional therapeutic agents include microtubule-binding agents, DNA intercalators or cross-linkers, DNA synthesis inhibitors, DNA and/or RNA transcription inhibitors, antibodies, enzymes, enzyme inhibitors, and gene regulators. These agents (which are administered at a therapeutically effective amount) and treatments can be used alone or in combination. Methods and therapeutic dosages of such agents are known to those skilled in the art, and can be determined by a skilled clinician.
  • Microtubule-binding agent refers to an agent that interacts with tubulin to stabilize or destabilize microtubule formation thereby inhibiting cell division.
  • microtubule-binding agents that can be used in conjunction with the disclosed therapy include, without limitation, paclitaxel, docetaxel, vinblastine, vindesine, vinorelbine (navelbine), the epothilones, colchicine, dolastatin 15, nocodazole, podophyllotoxin and rhizoxin. Analogs and derivatives of such compounds also can be used and are known to those of ordinary skill in the art. For example, suitable epothilones and epothilone analogs are described in International Publication No.
  • Taxoids such as paclitaxel and docetaxel, as well as the analogs of paclitaxel taught by U.S. Patent Nos. 6,610,860; 5,530,020; and/or 5,912,264 can be used.
  • DNA and/or RNA transcription regulators including, without limitation, actinomycin D, daunorubicin, doxorubicin and derivatives and analogs thereof also are suitable for use in combination with the disclosed therapies
  • DNA intercalators and cross-linking agents that can be administered to a subject include, without limitation, cisplatin, carboplatin, oxaliplatin, mitomycins, such as mitomycin C, bleomycin, chlorambucil, cyclophosphamide and derivatives and analogs thereof
  • DNA synthesis inhibitors suitable for use as therapeutic agents include, without limitation, methotrexate, 5-fluoro-5'-deoxyuridine, 5-fluorouracil and analogs thereof.
  • suitable enzyme inhibitors include, without limitation, camptothecin, etoposide, formestane, trichostatin and derivatives and analogs thereof.
  • alkylating agents include carmustine or lomustine.
  • compounds that affect gene regulation include agents that result in increased or decreased expression of one or more genes, such as raloxifene, 5- azacytidine, 5-aza-2'-deoxycytidine, tamoxifen, 4-hydroxytamoxifen, mifepristone and derivatives and analogs thereof; and kinase inhibitors include Gleevac, Iressa, and Tarceva that prevent phosphorylation and activation of growth factors.
  • anti -tumor agents for example anti -tumor agents, that may or may not fall under one or more of the classifications above, also are suitable for administration in combination with the disclosed therapies.
  • such agents include adriamycin, apigenin, rapamycin, zebularine, cimetidine, and derivatives and analogues thereof.
  • At least a portion of the tumor is surgically removed (for example via cryotherapy), irradiated, chemically treated (for example via chemoembolization) or combinations thereof, prior to administration of the disclosed therapies (such as administration of fenoterol, a fenoterol analogue or a combination thereof).
  • a subject can have at least a portion of the tumor surgically excised prior to administration of the disclosed therapies.
  • one or more chemotherapeutic agents are administered following treatment with a composition including fenoterol, a fenoterol analogue or a combination thereof.
  • [ 3 H]-Thymidine (70-90 Ci/mmol) was purchased from PerkinElmer Life and Analytical Sciences (Waltham, MA).
  • Eagle's Minimum Essential Medium (E-MEM) trypsin solution
  • PBS phosphate-buffered saline
  • FBS fetal bovine serum
  • L-glutamine (200 mM) 100X solutions of sodium pyruvate
  • 200 mM fetal bovine serum
  • penicillin/streptomycin a mixture of 10,000 units/ml penicillin and 10,000 ⁇ g/ml streptomycin
  • Quality Biological Gaithersburg, MD
  • WIN 55,212-2, AM251, and AM630 were purchased from Cayman Chemical (Ann Arbor, MI).
  • ICI 118,551 hydrochloride and (R)-isoproterenol were obtained from Sigma-Aldrich (St. Louis, MO). Phenylmethylsulfonyl fluoride (PMSF), benzamidine, leupeptin, pepstatin A, MgCl 2 , EDTA, Trizma-Hydrochloride (Tris-HCl), ( ⁇ ) -propranolol and minimal essential medium (MEM) were obtained from Sigma Aldrich (St. Louis, MO). Egg phosphatidylcholine lipids (PC) were obtained from Avanti Polar Lipids (Alabaster, AL).
  • ( ⁇ )-fenoterol was purchased from Sigma - Aldrich and [ 3 H]-( ⁇ )-fenoterol was acquired from Amersham Biosciences (Boston, MA).
  • the organic solvents n-hexane, 2-propanol and triethylamine were obtained as ultra pure HPLC grade solvents from Carlo Erba (Milan, Italy).
  • Fetal bovine serum and penicillin-streptomycin were purchased from Life Technologies (Gaithersburg, MD), and [ 125 I]- (i)-iodocyanopindolol (ICYP) was purchased from NEN Life Science Products, Inc. (Boston, MA).
  • PANC-1 cells were incubated with media containing vehicle (0.01% DMSO) or (R,S')-MNF (0- 10 ⁇ ) for 24h. The medium was removed, and cells were collected and processed for immunoblot analysis as recently described (Singh et al., Pharmacological Research 111, pages 757-766, 2016). All membrane-bound primary antibodies were detected with horseradish peroxidase-conjugated secondary antibodies (Santa Cruz Biotechnology, Dallas, TX) and visualized by enhanced chemiluminescence (ECL Plus, GE Healthcare, Piscataway, NT).
  • Quantification of the protein bands was performed by volume densitometry using ImageJ software (National Institutes of Health, Bethesda, MD) and normalization to ⁇ -actin or Lamin A/C.
  • Primary antibodies used in this study were raised against EGFR (sc-03) and ⁇ -catenin (sc- 7199) (Santa Cruz Biotechnology); PKM2 (ab38237) and ⁇ -actin (ab6276) (Abeam, Cambridge, MA). Three independent experiments were conducted on three separate days.
  • mice Female Balb/c nude mice (aged between 6-8 weeks, weight 18-20 g) were purchased from HFK Bioscience Co., Ltd. (Beijing, China) and maintained under pathogen-free conditions with a 12-hour light/12-hour dark cycle. Animals had free access to drinking water and were fed ad libitum with normal chow. Each mouse was inoculated subcutaneously at the right flank region with PANC-1 cells (5 x 10 6 ) in O. lmL of PBS for tumor development.
  • the treatments were started at Day 8 when the mean tumor size reached 139 ⁇ 34 mm 3 and the mice weighed 21.1 ⁇ 1.1 g.
  • mice received a single intraperitoneal (ip) injection 5 days per week for 3 treatment cycles of either vehicle (1% hydroxypropyl-P-cyclodextrin) (Control), 20mg-kg _1 (R,S')-MNF (Arm 2) or 40mg-kg _1 (R,S')-MNF (Arm 2).
  • the dosing volume was adjusted according to weight (lC ⁇ L/g). Tumor volumes and weights were determined at the beginning and end of each dosing cycle, while visual estimation of food and water consumption, eye/hair matting, and behavior such as mobility were determined daily.
  • the animals were euthanized by cervical extension 5 days after the end of the last dosing cycle.
  • Plasma samples were collected at the conclusion of the studies and, after collection of plasma samples, the tumors were excised, weighed, divided into three portions and snap frozen.
  • the xenograft studies were conducted at the Crown Biosciences, Ltd. facilities and all protocols were approved by the Animal Care and Use Committee at CrownBio (AN- 1407-009- 164), which are based on "the Guide for the Care and Use of Laboratory Animals” (NRC 2011).
  • Statistical analysis Sigmoidal dose-response curves (IC 50 curves) were determined using the 'nonlinear regression (curve fit)' model contained within the Prism 4 software package (GraphPad Software, Inc) running on a personal computer. For immunoblot analyses, tumor volume, and animal weight analyses, statistical comparisons between treated and control groups were performed using unpaired Student's t-tests. P values ⁇ 0.05 were considered significant.
  • mice Female Balb/c nude mice (aged between 6-8 weeks, weight 18-20 g) were purchased from HFK Bioscience Co., Ltd. (Beijing, China) and maintained under pathogen-free conditions with a 12-h light/12-h dark cycle. Animals had free access to drinking water and were fed ad libitum with normal chow. Each mouse was inoculated subcutaneously at the right flank region with PANC-1 cells (5- 106) in O. lmL of PBS for tumor development.
  • mice received an i.p. injection of either vehicle (20% hydroxypropyl-P-cyclodextrin) (Control) or 10 mg-kg "1 MNF once daily for 16 days adjusted to 25 mg-kg "1 for the last 5 days (Experimental).
  • Protein concentration in clarified lysates was determined using the bicinchoninic acid reagent (Thermo Fisher Scientific, Waltham, MA, USA). Proteins (20 ⁇ g/well) were separated on 4-12% precast gels (Invitrogen, Carlsbad, CA, USA) using SDS-polyacrylamide gel electrophoresis under reducing conditions and then electrophoretically transferred onto polyvinylidene fluoride membrane (Invitrogen). Western blots were performed according to standard methods, which involved a blocking step in Tris-buffered saline/0.1% Tween-20 (TBS-T) supplemented with 5% non-fat milk and incubation with primary antibodies of interest.
  • TBS-T Tris-buffered saline/0.1% Tween-20
  • the primary antibodies used in this study were raised against EGFR (sc-03), MCT4 (sc- 50329), Glut8 (sc-30108), and ⁇ -catenin (sc-7199) (Santa Cruz Biotechnology); PKM2 (ab38237) and ⁇ -actin (ab6276) (Abeam, Cambridge, MA, USA); hexokinase II (cat. #2867) and PDKl (cat. #3062S) (Cell Signaling Technology, Beverly, MA, USA). The antibodies were used at the dilution recommended by the manufacturers.
  • the mixture was centrifuged for 15 min at 14000 rpm at 4 °C and the supernatant collected and analyzed using a system composed of an Agilent Technologies 1100 LC/MSD equipped with a G1322A degasser, G1312A quaternary pump, G1367A autosampler, G1316A column thermostat and G1946D mass spectrometer supplied with electrospray ionization (ESI).
  • ESI electrospray ionization
  • SIM Selected ion monitoring
  • the compounds of interest were monitored in the positive-ion mode for SIM at m/z 162.1 (carnitine), m/z 90.1 (lactate) and m/z 105.1 (3-hydroxybutyrate).
  • the internal standard was monitored at m/z 195.1 (p-aminohippuric acid).
  • the C6 cell line is derived from a rat glioblastoma brain tumor and is a standard model for brain tumors.
  • (R,R')-M F decreases glucose uptake and L-lactate output in this cell line, expanding the effect to more than one type of tumor.
  • the human pancreatic tumor cell line, PANC-1, breast cancer cell lines MDA-MB-231 and MCF-7, and rat-derived C6 glioblastoma cell line were purchased from ATCC (Manassas, VA, USA). Upon receipt of the three cell lines, cells were expanded for a few passages to enable the generation of new frozen stocks. Cells were resuscitated as needed and used for fewer than 6 months after resuscitation (no more than 10 passages). ATCC performs thorough cell line authentication utilizing Short Tandem Repeat (STR) profiling.
  • STR Short Tandem Repeat
  • PANC-1 and C6 cells were maintained in DMEM with L-glutamine supplemented with 10% FBS and 1% penicillin/streptomycin.
  • MDA-MB-231 cells were maintained in RPMI- 1640 supplemented with 10% FBS and 1% penicillin/streptomycin and MCF-7 cells were maintained in EMEM with L-glutamine supplemented with 10% FBS and 0.01 mg-ml "1 human recombinant insulin.
  • Cells were maintained in a controlled environment (37 °C under humidified 5% C02 in air), and the medium was replaced every 2-3 days. Prior to experiments, cells were seeded on 100 x 20 mm tissue culture plates and grown to -70% confluency unless stated otherwise.
  • C6 cells Glucose consumption and lactate production in C6 cells.
  • C6 cells were culture in 48-well plates and treated with MNF (0.02 or 0.20 ⁇ ) or vehicle (DMSO, 0.1%) and glucose and lactate concentrations in the incubation media were periodically monitored for 48 h.
  • MNF 0.02 or 0.20 ⁇
  • DMSO vehicle
  • glucose and lactate concentrations in the incubation media were periodically monitored for 48 h.
  • the culture media were collected to determine glucose and lactate content using the Liquick Cor-Glucose and Liquick Cor-Lactate Diagnostic Kits (Cormay, Lublin, Poland). Measurements were carried out according to the manufacturer's protocols.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Emergency Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

Methods of regulating disorders and diseases by reducing tumor generated L-lactate in an environment surrounding a tumor, include administration of a fenoterol analogue, such as for example, MNF. The method may increase a cancer patient's response to an immune checkpoint blockade therapy.

Description

METHODS OF TREATING CANCER
TECHNICAL FIELD
[0001] The present disclosure relates to methods of treating cancer by reducing tumor generated L-lactate in the microenvironment surrounding a tumor by administration of at least one agent, such as for example a fenoterol analogue.
BACKGROUND
[0002] Cancer is the second leading cause of human death next to coronary disease in the
United States. Worldwide, millions of people die from cancer every year. In the United States alone, as reported by the American Cancer Society, cancer causes the death of well over a half- million people annually, with over 1.2 million new cases diagnosed per year. While deaths from heart disease have been declining significantly, those resulting from cancer generally are on the rise. Cancer is soon predicted to become the leading cause of death.
SUMMARY
[0003] This disclosure concerns the discovery that the response to immune checkpoint blockade therapies in cancer patients can be increased by reducing tumor generated L-lactate in the microenvironment surrounding a tumor. In embodiments, fenoterol analogues are used to improve the effectiveness of one or more immune checkpoint blockade therapy by reducing tumor generated L-lactate in the microenvironment surrounding a tumor. The exemplary methods described herein can be used, for example, to improve the effectiveness of an immune checkpoint blockade therapy in the treatment of pancreatic cancer, breast cancer, or other cancers. [0004] In embodiments, the method includes administering a therapeutically effective amount of a fenoterol analogue to a cancer patient to improve the effectiveness of one or more immune checkpoint blockade therapies. In embodiments, the fenoterol analogue is an antagonist of pyruvate kinase M2 (PKM2). In embodiments, the fenoterol analogue is an antagonist of hexokinase-2 (HK2). In embodiments, the fenoterol analogue is a compound that attenuates monocarboxylate transporter 4 (MCT4) expression and/or function, thereby decreasing L-lactate export and increasing L-lactate concentrations within the tumor.
[0005] In embodiments, fenoterol analogues include one or more compounds selected from the group consisting of (R,R')-4'-methoxy- 1-naphthylfenoterol ("MNF"), (R,S')-4'- methoxy- 1-naphthylfenoterol, (R,R')-ethylMNF, (R,R')-napthylfenoterol, (R,S napthylfenoterol, (R,R')-ethyl-naphthylfenoterol, (R,R')-4' -amino- 1-naphthylfenoterol, (R,R')- 4'-hydroxy-l-naphthylfenoterol, (R,R')-4-methoxy-ethylfenoterol, (R,R')-methoxyfenoterol, (R,R')- ethylfenoterol, (R,R')-fenoterol and their respective stereoisomers.
[0006] In embodiments, the fenoterol analogue is (R,R')-4'-methoxy- 1-naphthylfenoterol (MNF), a compound having the formula:
Figure imgf000003_0001
[0007] In embodiments, the presently described methods include administering a therapeutically effective amount of a pharmaceutical composition containing a fenoterol analogue and a pharmaceutically acceptable carrier to a cancer patient to decrease the amount of L-lactate in a tumor microenvironment. In embodiments, the cancer patient is known to have pancreatic or breast cancer. In embodiments, the method includes administering one or more therapeutic agents in addition to a fenoterol analogue. The methods can include administration of the one or more therapeutic agents separately, sequentially or concurrently, for example in a combined composition with a fenoterol analogue. In embodiments, the one or more therapeutic agents administered in addition to a fenoterol analogue may be one or more immune checkpoint blockade therapy.
[0008] In another aspect, a method of attenuating monocarboxylate transporter 4 (MCT4) expression and/or function is described. Attenuating MCT4 expression and/or function decreases L-lactate export out of cancer cells and increases L-lactate concentrations in cancer cells. In embodiments, the method includes administering a therapeutically effective amount of a fenoterol analogue to attenuate MCT4 expression and/or function.
[0009] In another aspect, a method of decreasing the amount (and hence activity) of hexokinase-2 (HK2) in cancer cells showing increased HK2 expression is described. Decreasing the amount HK2 decreases glucose metabolism and glycolysis and increases the sensitivity of cancer cells to cell death inducers. In embodiments, the method includes administering a therapeutically effective amount of a fenoterol analogue to decrease expression of HK2.
[0010] In another aspect, a method of decreasing the amount (and hence activity) of glucose transporter facilitators (e.g., Glut-1, Glut-8, and others) in cancer cells is described. In embodiments, the method includes administering a therapeutically effective amount of a fenoterol analogue to decrease expression of one or more glucose transporter facilitators.
[0011] The foregoing simplified summary of the claimed subject matter is presented in order to provide a basic understanding of some aspects of the claimed subject matter. The foregoing summary is not an extensive overview of the claimed subject matter. It is intended to neither identify key or critical elements of the claimed subject matter nor delineate the scope of the claimed subject matter. Its sole purpose is to present some concepts of the claimed subject matter in a simplified form as a prelude to the more detailed description that is presented hereinbelow. Further scope of applicability of the present disclosure will become apparent from the detailed description given hereinafter. However, it should be understood that the detailed description and specific examples, while indicating specific embodiments of the present disclosure, are given by way of illustration only, since various changes and modifications within the spirit and scope of the present disclosure will become apparent to those skilled in the art from this detailed description.
BRIEF DESCRIPTION OF THE DRAWINGS
[0012] Fig. 1 shows that (R,S')-MNF reduces plasma L-lactate concentrations in mice bearing PANC-1 tumor xenografts;
[0013] Fig. 2 shows that (R,S')-MNF dose-dependently reduces β-catenin expression in
PANC-1 tumor cells;
[0014] Fig. 3 shows that (R,R')-MNF inhibits glucose uptake and lactate production in
C6 glioma cells ;
[0015] Figure 4 shows that (R,R')-MNF reduces lactate production in the breast cancer cell lines MCF-7 and MDA-MB-231;
[0016] Figure 5 shows the pathway of aerobic glycolysis in cancer cells resulting in an enhanced uptake of glucose and production of L-lactate ("the Warburg effect");
[0017] Fig. 6 shows that (R,R')-MNF reduces the expression of HK2 and the L-lactate transporter MCT4 in tumor tissue obtained from the PANC1 xenograft; [0018] Figure 7 shows that (R,R')-MNF reduces the expression of β-catenin in tumor tissues obtained from mice bearing a PANC-1 xenograft;
[0019] Figure 8 shows that in mice with a PANC-1 xenograft tumor, (R,R')-MNF decreases the plasma concentration of L-lactate and increases the tumor L-lactate concentration; and
[0020] Figure 9 shows the impact of lactate on tumor microenvironment. .
DETAILED DESCRIPTION
Introduction
[0021] Cancer cells evolve several alterations in their metabolism to survive in unfavorable microenvironments, while retaining their ability to proliferate. In pancreatic cancer, metabolic reprogramming is a key aspect of tumorigenesis and has a profound effect on gene expression, cellular differentiation, metastasis, and tumor microenvironment. One metabolic adaptation of tumor cells is a shift to aerobic glycolysis as a main source of ATP, rather than oxidative phosphorylation (OXPHOS), irrespective of oxygen availability, a phenomenon referred to as the Warburg effect. This phenotype may promote a state of apoptosis resistance, the generation of biosynthetic precursors for proliferation, and increased invasive ability. Thus, aerobic glycolysis is a major component of metabolic reprograming and is characterized by enhanced glucose uptake and its conversion to L-lactate via the glycolytic pathway.
[0022] The dimeric form of pyruvate kinase M2 ("PKM2") is a key regulator of cancer metabolism, driving both lactate formation and upregulation of genes associated with glycolysis, tumor proliferation, and autoinduction of PKM2 expression. [0023] The glycolytic enzyme hexokinase 2 (HK2) may also impact the Warburg effect in cancer cells showing increased HK2 expression. Depletion of HK2 has been shown to restore oxidative glucose metabolism and increase sensitivity to cell death inducers such as radiation and chemotherapies.
[0024] Another key aspect of metabolic reprogramming is the MCT4-mediated export of
L-lactate. In pancreatic cancer, increased expression of MCT4 is associated with a poor prognosis due to the role that increased L-lactate concentration plays in immunoresistance. Tumor-generated L-lactate inhibits anticancer immune response through decreased cytotoxic activity of T lymphocytes and natural killer cells.
[0025] Thus, therapeutic strategies to modulate the Warburg effect may interfere with growth and therapeutic sensitivity of some cancer cells.
[0026] Incubation of pancreatic cancer cells (e.g., PANC-1 cells) with a fenoterol analog in accordance with the methods of the present disclosure attenuates glycolysis by reducing the expression of Glut-1, hexokinase II, PKM2, and lactate dehydrogenase. Administration of a fenoterol analog in accordance with the methods of the present disclosure reduces L-lactate plasma concentration relative to pre-dose concentrations. Since immunotherapy lacks therapeutic efficacy in pancreatic cancer and some of the observed resistance has been attributed to the tumor microenvironment, reduction of L-lactate concentration in the tumor microenvironment in accordance with the presently described methods potentiates the effect of checkpoint inhibitors and therapeutic vaccines.
[0027] In accordance with exemplary embodiments of the present disclosure, a PKM2 antagonist, such as a fenoterol analogue, is used to treat a disease state by reducing tumor generated L-lactate in the environment surrounding a tumor. This change in the microenvironment surrounding the tumor may reduce cellular proliferation, the expression of proteins key to the survival of cancer cells, and the resistance of cancer cells to treatment by anticancer drugs (known as multidrug resistance).
[0028] In accordance with other exemplary embodiments of the present disclosure, a compound that decreases HK2 expression and activity, such as a fenoterol analogue, is used to treat a disease state. In such embodiments, cancer is treated by restoring oxidative glucose metabolism and increasing sensitivity of cancer cells to cell death inducers.
[0029] In accordance with other exemplary embodiments of the present disclosure, a compound that attenuates MCT4 expression and/or function, such as a fenoterol analogue, is used to treat a disease state. In such embodiments, the MCT4-mediated export of L-lactate is reduced, thereby reducing tumor-generated L-lactate in the environment surrounding a tumor enhancing anticancer immune response and increasing cytotoxic activity of T lymphocytes and natural killer cells.
[0030] Thus, the compounds described herein can be used to treat pancreatic or breast cancer as well as other forms of cancer. Based upon these findings, methods of treating disorders and diseases modulated by glycolysis are described.
Abbreviations and Terms
[0031] Abbreviations:
AM251 : l-(2,4-dichlorophenyl)-5-(4-iodophenyl)-4-methyl-N-(l-piperidyl)pyrazole- 3- carboxamide AM630: l-[2-(morpholin-4-yl)ethyl]-2-methyl-3-(4-methoxybenzoyl)-6-iodoindole AR: adrenergic receptor
2-AR: 2-adrenergic receptor
CB: cannabinoid
EGFR: epidermal growth factor receptor
ERK: extracellular regulated kinase
GPR55: G protein-coupled receptor 55
GPCR: G protein-coupled receptor
HPLC: high performance liquid chromatography
ICI 118,551 : 3-(isopropylamino)-l-[(7-methyl-4-indanyl)oxy]butan-2-ol
ICYP: [125I]cyanopindolol
IP: intraperitoneal
IV: intravenous
MNF : (R,R' )-4-methoxy- 1 -naphthylfenoterol
NF: naphthylfenoterol
UV: ultraviolet
[0032] Terms:
[0033] Unless otherwise explained, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which the disclosed subject matter belongs. Definitions of common terms in chemistry may be found in The McGraw-Hill Dictionary of Chemical Terms, 1985, and The Condensed Chemical Dictionary, 1981.
[0034] Except as otherwise noted, any quantitative values are approximate whether the word "about" or "approximately" or the like are stated or not. The materials, methods, and examples described herein are illustrative only and not intended to be limiting. Any molecular weight or molecular mass values are approximate and are provided only for description. Except as otherwise noted, the methods and techniques of the present invention are generally performed according to conventional methods well known in the art and as described in various general and more specific references that are cited and discussed throughout the present specification. See, e.g., Loudon, Organic Chemistry, Fourth Edition, New York: Oxford University Press, 2002, pp. 360-361, 1084-1085; Smith and March, March' s Advanced Organic Chemistry: Reactions, Mechanisms, and Structure, Fifth Edition, Wiley- Interscience, 2001; or Vogel, A Textbook of Practical Organic Chemistry, Including Qualitative Organic Analysis, Fourth Edition, New York: Longman, 1978.
[0035] In order to facilitate review of the various embodiments disclosed herein, the following explanations of specific terms are provided:
[0036] Acyl: A group of the formula RC(O)- wherein R is an organic group.
[0037] Acyloxy: A group having the structure -OC(0)R, where R may be an optionally substituted alkyl or optionally substituted aryl. "Lower acyloxy" groups are those where R contains from 1 to 10 (such as from 1 to 6) carbon atoms. [0038] Administration: To provide or give a subject a composition, such as a pharmaceutical composition including one or more fenoterol analogues by any effective route. Exemplary routes of administration include, but are not limited to, injection (such as subcutaneous, intramuscular, intradermal, intraperitoneal ("IP"), and intravenous ("IV")), oral, sublingual, rectal, transdermal, intranasal, vaginal and inhalation routes.
[0039] Alkoxy: A radical (or substituent) having the structure -O-R, where R is a substituted or unsubstituted alkyl. Methoxy (-OCH3) is an exemplary alkoxy group. In a substituted alkoxy, R is alkyl substituted with a non-interfering substituent. "Thioalkoxy" refers to -S-R, where R is substituted or unsubstituted alkyl. "Haloalkyloxy" means a radical -OR where R is a haloalkyl.
[0040] Alkoxy carbonyl: A group of the formula -C(0)OR, where R may be an optionally substituted alkyl or optionally substituted aryl. "Lower alkoxy carbonyl" groups are those where R contains from 1 to 10 (such as from 1 to 6) carbon atoms.
[0041] Alkyl: An acyclic, saturated, branched- or straight-chain hydrocarbon radical, which, unless expressly stated otherwise, contains from one to fifteen carbon atoms; for example, from one to ten, from one to six, or from one to four carbon atoms. This term includes, for example, groups such as methyl, ethyl, n-propyl, isopropyl, isobutyl, t-butyl, pentyl, heptyl, octyl, nonyl, decyl, or dodecyl. The term "lower alkyl" refers to an alkyl group containing from one to ten carbon atoms. Unless expressly referred to as an "unsubstituted alkyl," alkyl groups can either be unsubstituted or substituted. An alkyl group can be substituted with one or more substituents (for example, up to two substituents for each methylene carbon in an alkyl chain). Exemplary alkyl substituents include, for instance, amino groups, amide, sulfonamide, halogen, cyano, carboxy, hydroxy, mercapto, trifluorom ethyl, alkyl, alkoxy (such as methoxy), alkylthio, thioalkoxy, arylalkyl, heteroaryl, alkylamino, dialkylamino, alkylsulfano, keto, or other functionality.
[0042] Amino carbonyl (carbamoyl): A group of the formula C(0)N(R)R', wherein R and R are independently of each other hydrogen or a lower alkyl group.
[0043] p2-adrenergic receptor (P2-AR): A subtype of adrenergic receptors that are members of the G-protein coupled receptor family. P2-AR subtype is involved in respiratory diseases, cardiovascular diseases, premature labor and, as disclosed herein, tumor development. Increased expression of p2-ARs can serve as therapeutic targets.
[0044] Cannabinoid Receptors: A class of cell membrane receptors under the G protein- coupled receptor superfamily. The cannabinoid receptors contain seven transmembrane spanning domains. Cannabinoid receptors are activated by three major groups of ligands, endocannabinoids (produced by the mammalian body), plant cannabinoids (such as THC, produced by the cannabis plant) and synthetic cannabinoids (such as HU-210). All of the endocannabinoids and plant cannabinoids are lipophilic, i.e., fat soluble, compounds. Two subtypes of cannabinoid receptors are CBi (see GenBank Accession No. NM_033181 mRNA and UniProt P21554, each of which is hereby incorporated by reference as of May 23, 2012) and CB2 (see GenBank Accession No. NM_001841 mRNA and UniProt P34972, each of which is hereby incorporated by reference as of May 23, 2012). The CB2 receptor is expressed mainly in the immune system and in hematopoietic cells. Additional non-CBi and non-CB2 include GPR55 (GenBank Accession No. NM_005683.3 or NP_005674.2 protein, each of which is hereby incorporated by reference as of May 23, 2012), GPR119 (GenBank Accession No. NM_178471.2 or NP_848566.1 protein, each of which is hereby incorporated by reference as of May 23, 2012) and GPR18 (also known as N- arachidonyl glycine receptor and involved in microglial migration, GenBank Accession No. NM_001098200 mRNA, NP_001091670.1, each of which is hereby incorporated by reference as of May 23, 2012).
[0045] The protein sequences of CBi and CB2 receptors are about 44% similar. When only the transmembrane regions of the receptors are considered, amino acid similarity between the two receptor subtypes is approximately 68%. In addition, minor variations in each receptor have been identified. Cannabinoids bind reversibly and stereo-selectively to the cannabinoid receptors. The affinity of an individual cannabinoid to each receptor determines the effect of that cannabinoid. Cannabinoids that bind more selectively to certain receptors are more desirable for medical usage. GPR55 is coupled to the G-protein Gi3 and/or Gn and activation of the receptor leads to stimulation of rhoA, cdc42 and rack GPR55 is activated by the plant cannabinoids A9- THC and cannabidiol, and the endocannabinoids anandamide, 2-AG, noladin ether in the low nanomolar range. In contrast, CBi and CB2 receptors are coupled to inhibitory G proteins. This indicates that both types of receptors will have different readouts. For example, activation of CBi causes apoptosis whereas increase in GPR55 activity is oncogenic. The CBi receptor antagonist (also termed 'inverse agonist') compound, AM251, is, in fact, an agonist for GPR55. It binds GPR55 and is readily internalized. This illustrates the opposite behavior of these two GPCRs.
[0046] Carbamate: A group of the formula -OC(0)N(R)-, wherein R is H, or an aliphatic group, such as a lower alkyl group or an aralkyl group. [0047] Chemotherapy; chemotherapeutic agents: As used herein, any chemical agent with therapeutic usefulness in the treatment of diseases characterized by abnormal cell growth. Such diseases include tumors, neoplasms, and cancer as well as diseases characterized by hyperplastic growth. In one embodiment, a chemotherapeutic agent is an agent of use in treating neoplasms such as solid tumors, including a tumor associated with CB receptor activity and/or expression. In embodiments, a chemotherapeutic agent is radioactive molecule. In embodiments, a CB receptor regulator, such as one or more fenoterol analogues or a combination thereof is a chemotherapeutic agent. In one example, a chemotherapeutic agent is carmustine, lomustine, procarbazine, streptozocin, or a combination thereof. One of skill in the art can readily identify a chemotherapeutic agent of use (e.g., see Slapak and Kufe, Principles of Cancer Therapy, Chapter 86 in Harrison's Principles of Internal Medicine, 14th edition; Perry et al., Chemotherapy, Ch. 17 in Abel off, Clinical Oncology 2nd ed., © 2000 Churchill Livingstone, Inc; Baltzer L., Berkery R. (eds): Oncology Pocket Guide to Chemotherapy, 2nd ed. St. Louis, Mosby-Year Book, 1995; Fischer DS, Knobf MF, Durivage HJ (eds): The Cancer Chemotherapy Handbook, 4th ed. St. Louis, Mosby-Year Book, 1993).
[0048] Control or Reference Value: A "control" refers to a sample or standard used for comparison with a test sample. In some embodiments, the control is a sample obtained from a healthy subject or a tissue sample obtained from a patient diagnosed with a disorder or disease, such as a tumor, that did not respond to treatment with a p2-agonist. In some embodiments, the control is a historical control or standard reference value or range of values.
[0049] Derivative: A chemical substance that differs from another chemical substance by one or more functional groups. In embodiments, a derivative retains a biological activity of a molecule from which it was derived. [0050] Effective amount: An amount of agent that is sufficient to generate a desired response, such as reducing or inhibiting one or more signs or symptoms associated with a condition or disease. When administered to a subject, a dosage will generally be used that will achieve target tissue concentrations. In some examples, an "effective amount" is one that treats one or more symptoms and/or underlying causes of any of a disorder or disease. In some examples, an "effective amount" is a "therapeutically effective amount" in which the agent alone with an additional therapeutic agent(s) (for example a chemotherapeutic agent) induces the desired response such as treatment of a tumor. In one example, a desired response is to decrease tumor size or metastasis in a subject to whom the therapy is administered. Tumor metastasis does not need to be completely eliminated for the composition to be effective. For example, a composition can decrease metastasis by a desired amount, for example by at least 20%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, at least 98%, or even at least 100%) (elimination of the tumor), as compared to metastasis in the absence of the composition.
[0051] In particular examples, it is an amount of an agent effective to decrease a number of carcinoma cells, such as in a subject to whom it is administered, for example a subject having one or more carcinomas. The cancer cells do not need to be completely eliminated for the composition to be effective. For example, a composition can decrease the number of cancer cells by a desired amount, for example by at least 20%, at least 50%, at least 60%>, at least 70%, at least 80%), at least 90%, at least 95%, at least 98%, or even at least 100% (elimination of detectable cancer cells), as compared to the number of cancer cells in the absence of the composition. [0052] The effective amount of a composition useful for reducing, inhibiting, and/or treating a disorder in a subject will be dependent on the subject being treated, the severity of the disorder, and the manner of administration of the therapeutic composition. Effective amounts a therapeutic agent can be determined in many different ways, such as assaying for a reduction in tumor size or improvement of physiological condition of a subject having a tumor, such as a brain tumor. Effective amounts also can be determined through various in vitro, in vivo or in situ assays.
[0053] Fenoterol Analogues: Fenoterol analogues include (R,R')-4'-methoxy-l- naphthylfenoterol ("MNF"), (R,S')-4'-methoxy-l-naphthylfenoterol, (R,R')-ethylMNF, (R,R')- napthylfenoterol, (R,S')-napthylfenoterol, (R,R')-ethyl-naphthylfenoterol, (R,R' )-4' -amino- 1- naphthylfenoterol, (R,R')-4'-hydroxy-l-naphthylfenoterol, (R,R')-4'-methoxy-ethylfenoterol, (R,R')-methoxyfenoterol, (R,R')-ethylfenoterol, (R,R')-fenoterol and their respective stereoisomers.
[0054] Inflammation: When damage to tissue occurs, the body's response to the damage is usually inflammation. The damage may be due to trauma, lack of blood supply, hemorrhage, autoimmune attack, transplanted exogenous tissue or infection. This generalized response by the body includes the release of many components of the immune system (for instance, IL-1 and TNF), attraction of cells to the site of the damage, swelling of tissue due to the release of fluid and other processes.
[0055] Isomers: Compounds that have the same molecular formula but differ in the nature or sequence of bonding of their atoms or the arrangement of their atoms in space are termed "isomers". Isomers that differ in the arrangement of their atoms in space are termed "stereoisomers". Stereoisomers that contain two or more chiral centers and are not mirror images of one another are termed "diastereomers." Steroisomers that are non-superimposable mirror images of each other are termed "enantiomers." When a compound has an asymmetric center, for example, if a carbon atom is bonded to four different groups, a pair of enantiomers is possible. An enantiomer can be characterized by the absolute configuration of its asymmetric center and is described by the R- and S-sequencing rules of Cahn and Prelog, or by the manner in which the molecule rotates the plane of polarized light and designated as dextrorotatory or levorotatory (i.e., as (+) or (-) isomers, respectively). A chiral compound can exist as either an individual enantiomer or as a mixture thereof. A mixture containing equal proportions of the enantiomers is called a "racemic mixture."
[0056] The compounds described herein may possess one or more asymmetric centers; such compounds can therefore be produced as individual (R), (S), (R,R), (R,S'), (S,R') and (S,S')-stereoisomers or as mixtures thereof. Unless indicated otherwise, the description or naming of a particular compound in the specification and claims is intended to include both individual enantiomers and mixtures, racemic or otherwise, thereof. The methods for the determination of stereochemistry and the separation of stereoisomers are well known in the art (see, e.g., March, Advanced Organic Chemistry, 4th edition, New York: John Wiley and Sons, 1992, Chapter 4).
[0057] Optional: "Optional" or "optionally" means that the subsequently described event or circumstance can but need not occur, and that the description includes instances where said event or circumstance occurs and instances where it does not. [0058] Pharmaceutically Acceptable Carriers: The pharmaceutically acceptable carriers
(vehicles) useful in this disclosure are conventional. Remington's Pharmaceutical Sciences, by E. W. Martin, Mack Publishing Co., Easton, PA, 19th Edition (1995), describes compositions and formulations suitable for pharmaceutical delivery of one or more therapeutic compounds or molecules, such as one or more nucleic acid molecules, proteins or antibodies that bind these proteins, and additional pharmaceutical agents.
[0059] In general, the nature of the carrier will depend on the particular mode of administration being employed. For instance, parenteral formulations usually comprise injectable fluids that include pharmaceutically and physiologically acceptable fluids such as water, physiological saline, balanced salt solutions, aqueous dextrose, glycerol or the like as a vehicle. For solid compositions (for example, powder, pill, tablet, or capsule forms), conventional non-toxic solid carriers can include, for example, pharmaceutical grades of mannitol, lactose, starch, or magnesium stearate. In addition to biologically-neutral carriers, pharmaceutical compositions to be administered can contain minor amounts of non-toxic auxiliary substances, such as wetting or emulsifying agents, preservatives, and pH buffering agents and the like, for example sodium acetate or sorbitan monolaurate.
[0060] Phenyl: Phenyl groups may be unsubstituted or substituted with one, two or three substituents, with substituent(s) independently selected from alkyl, heteroalkyl, aliphatic, heteroaliphatic, thioalkoxy, halo, haloalkyl (such as -CF3), nitro, cyano, -OR (where R is hydrogen or alkyl), -N(R)R (where R and R are independently of each other hydrogen or alkyl), -COOR (where R is hydrogen or alkyl) or -C(0)N(R)R" (where R and R" are independently selected from hydrogen or alkyl). [0061] Purified: The term "purified" does not require absolute purity; rather, it is intended as a relative term. Thus, for example, a purified preparation is one in which a desired component such as an (R,R ')-enantiomer of fenoterol is more enriched than it was in a preceding environment such as in a (+)-fenoterol mixture. A desired component such as (R,R')-enantiomer of fenoterol is considered to be purified, for example, when at least about 70%, 80%, 85%, 90%, 92%), 95%), 97%), 98%), or 99% of a sample by weight is composed of the desired component. Purity of a compound may be determined, for example, by high performance liquid chromatography (HPLC) or other conventional methods. In an example, the fenoterol analogue enantiomers are purified to represent greater than 90%, often greater than 95% of the other enantiomers present in a purified preparation. In other cases, the purified preparation may be essentially homogeneous, wherein other stereoisomers are less than 1%.
[0062] Compounds described herein may be obtained in a purified form or purified by any of the means known in the art, including silica gel and/or alumina chromatography. See, e.g., Introduction to Modern Liquid Chromatography, 2nd Edition, ed. by Snyder and Kirkland, New York: John Wiley and Sons, 1979; and Thin Layer Chromatography, ed. by Stahl, New York: Springer Verlag, 1969. In an example, a compound includes purified fenoterol or fenoterol analogue with a purity of at least about 70%, 80%, 85%, 90%, 92%, 95%, 97%, 98%, or 99% of a sample by weight relative to other contaminants. In a further example, a compound includes at least two purified stereoisomers each with a purity of at least about 70%, 80%, 85%, 90%, 92%, 95%), 97%), 98%), or 99% of a sample by weight relative to other contaminants. For instance, a compound can include a substantially purified (R,R')-fenoterol analogue and a substantially purified (R,S')-fenoterol analogue. [0063] Subject: The term "subject" includes both human and veterinary subjects, for example, humans, non-human primates, dogs, cats, horses, rats, mice, and cows. Similarly, the term mammal includes both human and non-human mammals.
[0064] Tissue: A plurality of functionally related cells. A tissue can be a suspension, a semi-solid, or solid. Tissue includes cells collected from a subject such as the brain or a portion thereof.
[0065] Tumor: All neoplastic cell growth and proliferation, whether malignant or benign, and all pre-cancerous and cancerous cells and tissues. A primary tumor is tumor growing at the anatomical site where tumor progression began and proceeded to yield this mass.
[0066] Under conditions sufficient for: A phrase that is used to describe any environment that permits the desired activity. In one example, under conditions sufficient for includes administering one or more fenoterol analogues to a subject to at a concentration sufficient to allow the desired activity. In some examples, the desired activity is reducing or inhibiting a sign or symptom associated with a disorder or disease, such as a breast or pancreatic, can be evidenced, for example, by a delayed onset of clinical symptoms of the tumor in a susceptible subject, a reduction in severity of some or all clinical symptoms of the tumor, a slower progression of the tumor (for example by prolonging the life of a subject having the tumor), a reduction in the number of tumor reoccurrence, an improvement in the overall health or well-being of the subject, or by other parameters well known in the art that are specific to the particular disease. In one particulate example, the desired activity is preventing or inhibiting tumor growth, such as breast cancer or pancreatic cancer growth. Tumor growth does not need to be completely inhibited for the treatment to be considered effective. For example, a partial reduction or slowing of growth such as at least about a 10% reduction, such as at least 20%, at least about 30%, at least about 40%, at least about 50% or greater is considered to be effective.
Chemical Structure of fenoterol analogues
[0067] Fenoterol analogues useful in the methods herein include (R,R')-4'-methoxy-l- naphthylfenoterol ("MNF"), (R,S')-4'-methoxy-l-naphthylfenoterol, (R,R')-ethylMNF, (R,R')- napthylfenoterol, (R,S')-napthylfenoterol, (R,R')-ethyl-naphthylfenoterol, (R,R' )-4' -amino- 1- naphthylfenoterol, (R,R')-4'-hydroxy-l-naphthylfenoterol, (R,R')-4'-methoxy-ethylfenoterol, (R,R')-4'-methoxyfenoterol, (R,R')-ethylfenoterol, (R,R')-fenoterol and their respective stereoisomers.
[0068] Examples of suitable groups for R1-R3 that can be cleaved in vivo to provide a hydroxy group include, without limitation, acyl, acyloxy and alkoxy carbonyl groups. Compounds having such cleavable groups are referred to as "prodrugs." The term "prodrug," as used herein, means a compound that includes a substituent that is convertible in vivo (e.g., by hydrolysis) to a hydroxyl group. Various forms of prodrugs are known in the art, for example, as discussed in Bundgaard, (ed.), Design of Prodrugs, Elsevier (1985); Widder, et al. (ed.), Methods in Enzymology, Vol. 4, Academic Press (1985); Krogsgaard-Larsen, et al., (ed), Design and Application of Prodrugs, Textbook of Drug Design and Development, Chapter 5, 113 191 (1991); Bundgaard, et al., Journal of Drug Delivery Reviews, 8: 1 38(1992); Bundgaard, Pharmaceutical Sciences, 77:285 et seq. (1988); and Higuchi and Stella (eds.) Prodrugs as Novel Drug Delivery Systems, American Chemical Society (1975). [0069] In embodiments, administering comprises administering a therapeutically effective amount of MNF, NF or a combination thereof. In some embodiments, administering comprises administering a therapeutically effective amount of MNF.
[0070] Particular method embodiments contemplate the use of solvates (such as hydrates), pharmaceutically acceptable salts and/or different physical forms of the fenoterol analogues herein described.
Solvates, Salts and Physical Forms
[0071] "Solvate" means a physical association of a compound with one or more solvent molecules. This physical association involves varying degrees of ionic and covalent bonding, including by way of example covalent adducts and hydrogen bonded solvates. In certain instances the solvate will be capable of isolation, for example when one or more solvent molecules are incorporated in the crystal lattice of the crystalline solid. "Solvate" encompasses both solution-phase and isolable solvates. Representative solvates include ethanol- associated compound, methanol-associated compounds, and the like. "Hydrate" is a solvate wherein the solvent molecule(s) is/are H20.
[0072] The disclosed compounds also encompass salts including, if several salt-forming groups are present, mixed salts and/or internal salts. The salts are generally pharmaceutically acceptable salts that are non-toxic. Salts may be of any type (both organic and inorganic), such as fumarates, hydrobromides, hydrochlorides, sulfates and phosphates. In an example, salts include non-metals (e.g., halogens) that form group VII in the periodic table of elements. For example, compounds may be provided as a hydrobromide salt. [0073] Additional examples of salt-forming groups include, but are not limited to, a carboxyl group, a phosphonic acid group or a boronic acid group, that can form salts with suitable bases. These salts can include, for example, nontoxic metal cations, which are derived from metals of groups IA, IB, IIA and IIB of the periodic table of the elements. In one embodiment, alkali metal cations such as lithium, sodium or potassium ions, or alkaline earth metal cations such as magnesium or calcium ions can be used. The salt can also be a zinc or an ammonium cation. The salt can also be formed with suitable organic amines, such as unsubstituted or hydroxyl -substituted mono-, di- or tri-alkylamines, in particular mono-, di- or tri-alkylamines, or with quaternary ammonium compounds, for example with N-methyl-N- ethylamine, diethylamine, triethylamine, mono-, bis- or tris- (2-hydroxy-lower alkyl)amines, such as mono-, bis- or tris- (2- hydroxyethyl)amine, 2-hydroxy-tert-butylamine or tris(hydroxymethyl)methylamine, N,N-di-lower alkyl-N-(hydroxy-lower alkyl)amines, such as N,N-dimethyl-N-(2-hydroxyethyl)amine or tri-(2- hydroxyethyl)amine, or N-methyl-D- glucamine, or quaternary ammonium compounds such as tetrabutylammonium salts.
[0074] Exemplary compounds disclosed herein possess at least one basic group that can form acid- base salts with inorganic acids. Examples of basic groups include, but are not limited to, an amino group or imino group. Examples of inorganic acids that can form salts with such basic groups include, but are not limited to, mineral acids such as hydrochloric acid, hydrobromic acid, sulfuric acid or phosphoric acid. Basic groups also can form salts with organic carboxylic acids, sulfonic acids, sulfo acids or phospho acids or N-substituted sulfamic acid, for example acetic acid, propionic acid, glycolic acid, succinic acid, maleic acid, hydroxymaleic acid, methylmaleic acid, fumaric acid, malic acid, tartaric acid, gluconic acid, glucaric acid, glucuronic acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, salicylic acid, 4-aminosalicylic acid, 2- phenoxybenzoic acid, 2-acetoxybenzoic acid, embonic acid, nicotinic acid or isonicotinic acid, and, in addition, with amino acids, for example with a-amino acids, and also with methanesulfonic acid, ethanesulfonic acid, 2-hydroxymethanesulfonic acid, ethane- 1,2-disulfonic acid, benzenedisulfonic acid, 4-methylbenzenesulfonic acid, naphthalene- 2-sulfonic acid, 2- or 3-phosphoglycerate, glucose-6-phosphate or N-cyclohexylsulfamic acid (with formation of the cyclamates) or with other acidic organic compounds, such as ascorbic acid. In a currently preferred embodiment, fenoterol is provided as a hydrobromide salt and exemplary fenoterol analogues are provided as their fumarate salts.
[0075] Additional counterions for forming pharmaceutically acceptable salts are found in
Remington's Pharmaceutical Sciences, 19th Edition, Mack Publishing Company, Easton, PA, 1995. In one aspect, employing a pharmaceutically acceptable salt may also serve to adjust the osmotic pressure of a composition.
[0076] In certain embodiments the compounds used in the method are provided are polymorphous. As such, the compounds can be provided in two or more physical forms, such as different crystal forms, crystalline, liquid crystalline or non-crystalline (amorphous) forms.
Use for the Manufacture of a Medicament
[0077] Any of the above described compounds (e.g., (R,R') and/or (R,S') fenoterol analogues or a hydrate or pharmaceutically acceptable salt thereof) or combinations thereof are intended for use in the manufacture of a medicament for treatment of breast or pancreatic cancer.
[0078] Formulations suitable for such medicaments, subjects who may benefit from same and other related features are described elsewhere herein.
Methods of Synthesis [0079] The disclosed fenoterol analogues can be synthesized by any method known in the art including those described in U.S. Patent Application Publication No. US 2010-0168245 Al, U.S. Patent Application Publication No. US 2012-0157543 Al and International Patent Publication No. WO 2011/112867, each of which is hereby incorporated by reference in its entirety. Many general references providing commonly known chemical synthetic schemes and conditions useful for synthesizing the disclosed compounds are available (see, e.g., Smith and March, March's Advanced Organic Chemistry: Reactions, Mechanisms, and Structure, Fifth Edition, Wiley- Interscience, 2001; or Vogel, A Textbook of Practical Organic Chemistry, Including Qualitative Organic Analysis, Fourth Edition, New York: Longman, 1978).
[0080] Compounds as described herein may be purified by any of the means known in the art, including chromatographic means, such as UPLC (including chiral UPLC), preparative thin layer chromatography, flash column chromatography and ion exchange chromatography. Any suitable stationary phase can be used, including normal and reversed phases as well as ionic resins. Most typically the disclosed compounds are purified via open column chromatography or prep chromatography.
[0081] Suitable exemplary syntheses of fenoterol analogues are provided below:
[0082] Scheme I: An exemplary synthesis of 4 stereoisomers of 1 - 6 including the coupling of the epoxide formed from either (R)- or (S)-3',5'-dibenzyloxyphenyl bromohydrin with the (R)- or (S)- enantiomer of the appropriate benzyl -protected 2-amino-3-benzylpropane (1 - 5) or the (R)- or (S)- enantiomer of N-benzyl-2-aminoheptane (6).
Figure imgf000026_0001
[0083] Scheme II: Exemplary synthesis of (R)-7 and (S)-7 using 2-phenethylamine. resulting compounds may be deprotected by hydrogenation over Pd/C and purified as fumarate salts.
Figure imgf000027_0001
[0084] Scheme III describes an exemplary synthesis for the chiral building blocks used in
Scheme II. The (R)- and (S)-3',5'-dibenzyloxyphenyl-bromohydrin enantiomers were obtained by the enantio specific reduction of 3,5-dibenzyloxy-a-bromoacetophenone using boron-methyl sulfide complex (BH3SCH3) and either (1R,2S)- or (1 S,2R)- cis-l-amino-2-indanol. The required (R)- and (S)-2-benzylaminopropanes were prepared by enantioselective crystallization of the rac-2-benzylaminopropanes using either (R)- or (S)-mandelic acid as the counter ion.
Figure imgf000028_0001
Pharmaceutical Compositions
[0085] The disclosed fenoterol analogues can be useful, at least, for reducing or inhibiting one or more symptoms or signs associated with cancer. Accordingly, pharmaceutical compositions comprising at least one disclosed fenoterol analogue are also described herein.
[0086] Formulations for pharmaceutical compositions are well known in the art. For example, Remington's Pharmaceutical Sciences, by E. W. Martin, Mack Publishing Co., Easton, PA, 19th Edition, 1995, describes exemplary formulations (and components thereof) suitable for pharmaceutical delivery of (R,R')-fenoterol and disclosed fenoterol analogues. Pharmaceutical compositions comprising at least one of these compounds can be formulated for use in human or veterinary medicine. Particular formulations of a disclosed pharmaceutical composition may depend, for example, on the mode of administration (e.g., oral or parenteral) and/or on the disorder to be treated. In some embodiments, formulations include a pharmaceutically acceptable carrier in addition to at least one active ingredient, such as a fenoterol compound.
[0087] Pharmaceutically acceptable carriers useful for the disclosed methods and compositions are conventional in the art. The nature of a pharmaceutical carrier will depend on the particular mode of administration being employed. For example, parenteral formulations usually comprise injectable fluids that include pharmaceutically and physiologically acceptable fluids such as water, physiological saline, balanced salt solutions, aqueous dextrose, glycerol or the like as a vehicle.
[0088] For solid compositions such as powder, pill, tablet, or capsule forms conventional non-toxic solid carriers can include, for example, pharmaceutical grades of mannitol, lactose, starch, or magnesium stearate. In addition to biologically neutral carriers, pharmaceutical compositions to be administered can optionally contain minor amounts of non-toxic auxiliary substances or excipients, such as wetting or emulsifying agents, preservatives, and pH buffering agents and the like; for example, sodium acetate or sorbitan monolaurate. Other non-limiting excipients include, nonionic solubilizers, such as cremophor, or proteins, such as human serum albumin or plasma preparations.
[0089] The disclosed pharmaceutical compositions may be formulated as a pharmaceutically acceptable salt. Pharmaceutically acceptable salts are non-toxic salts of a free base form of a compound that possesses the desired pharmacological activity of the free base. These salts may be derived from inorganic or organic acids. Non-limiting examples of suitable inorganic acids are hydrochloric acid, nitric acid, hydrobromic acid, sulfuric acid, hydriodic acid, and phosphoric acid. Non-limiting examples of suitable organic acids are acetic acid, propionic acid, glycolic acid, lactic acid, pyruvic acid, malonic acid, succinic acid, malic acid, maleic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, methyl sulfonic acid, salicylic acid, formic acid, trichloroacetic acid, trifluoroacetic acid, gluconic acid, asparagic acid, aspartic acid, benzenesulfonic acid, p- toluenesulfonic acid, naphthalenesulfonic acid, and the like. Lists of other suitable pharmaceutically acceptable salts are found in Remington's Pharmaceutical Sciences, 19th Edition, Mack Publishing Company, Easton, PA, 1995. A pharmaceutically acceptable salt may also serve to adjust the osmotic pressure of the composition.
[0090] The dosage form of a disclosed pharmaceutical composition will be determined by the mode of administration chosen. For example, in addition to injectable fluids, oral dosage forms may be employed. Oral formulations may be liquid such as syrups, solutions or suspensions or solid such as powders, pills, tablets, or capsules. Methods of preparing such dosage forms are known, or will be apparent, to those skilled in the art.
[0091] Certain embodiments of the pharmaceutical compositions comprising a disclosed compound may be formulated in unit dosage form suitable for individual administration of precise dosages. The amount of active ingredient such as (R,R')-MNF or F administered will depend on the subject being treated, the severity of the disorder, and the manner of administration, and is known to those skilled in the art. Within these bounds, the formulation to be administered will contain a quantity of the extracts or compounds disclosed herein in an amount effective to achieve the desired effect in the subject being treated.
[0092] In particular examples, for oral administration the compositions are provided in the form of a tablet containing from about 1.0 to about 50 mg of the active ingredient, particularly about 2.0 mg, about 2.5 mg, 5 mg, about 10 mg, or about 50 mg of the active ingredient for the symptomatic adjustment of the dosage to the subject being treated. In one exemplary oral dosage regimen, a tablet containing from about 1 mg to about 50 mg (such as about 2 mg to about 10 mg) active ingredient is administered two to four times a day, such as two times, three times or four times.
[0093] In other examples, a suitable dose for parental administration is about 1 milligram per kilogram (mg/kg) to about 100 mg/kg, such as a dose of about 10 mg/kg to about 80 mg/kg, such including about 1 mg/kg, about 2 mg/kg, about 5 mg/kg, about 20 mg/kg, about 30 mg/kg, about 40 mg/kg, about 50 mg/kg, about 80 mg/kg or about 100 mg/kg administered parenterally. However, other higher or lower dosages also could be used, such as from about 0.001 mg/kg to about 1 g/kg, such as about 0.1 to about 500 mg/kg, including about 0.5 mg/kg to about 200 mg/kg.
[0094] Single or multiple administrations of the composition comprising one or more of the disclosed compositions can be carried out with dose levels and pattern being selected by the treating physician. Generally, multiple doses are administered. In a particular example, the composition is administered parenterally once per day. However, the composition can be administered twice per day, three times per day, four times per day, six times per day, every other day, twice a week, weekly, or monthly. Treatment will typically continue for at least a month, more often for two or three months, sometimes for six months or a year, and may even continue indefinitely, i.e., chronically. Repeat courses of treatment are also possible.
[0095] In embodiments, the pharmaceutical composition is administered without concurrent administration of a second agent for the treatment of breast or pancreatic cancer. In one specific, non-limiting example, one or more of the disclosed compositions is administered without concurrent administration of other agents, such as without concurrent administration of an additional agent also known to target the tumor. In other specific non-limiting examples, a therapeutically effective amount of a disclosed pharmaceutical composition is administered concurrently with an additional agent, including an additional therapy. For example, the disclosed compounds are administered in combination with a chemotherapeutic agent, antioxidants, anti-inflammatory drugs or combinations thereof.
[0096] In embodiments, the disclosed compounds are administered in combination with an immune checkpoint therapy. Immune checkpoints affect immune system functioning and can be stimulatory or inhibitory. Tumors can use these checkpoints to protect themselves from immune system attacks. Checkpoint therapy can block inhibitory checkpoints, restoring immune system function. For example, the immune checkpoint therapy may impact the interaction between the transmembrane programmed cell death 1 protein (PDCDl, PD-1; also known as CD279) and its ligand, PD-1 ligand 1 (PD-L1, CD274). Cancer-mediated upregulation of PD-L1 on the cell surface may inhibit T cells that might otherwise attack. Thus, the presently disclosed fenoterol analogues may be administered with antibodies that bind to either PD-1 or PD-L1 and therefore block the interaction and thereby allow the T-cells to attack the tumor. In embodiments, the presently disclosed fenoterol analogues may be administered with IgG4 PD1 antibody (such as, for example, antibody BGB-A317, Nivolumab or Pembrolizumab which may provide benefits with respect to one or more of melanoma, lung cancer, kidney cancer and Hodgkin's lymphoma). In embodiments, the presently disclosed fenoterol analogues may be administered with a PD-L1 inhibitor, such as, for example, atezolizumab, avelumab, or and durvalumab. Alternatively or additionally, the presently disclosed fenoterol analogues may be administered with antibodies that block the immune checkpoint molecule CTLA-4, (such as, for example, ipilimumab, which may provide benefits with respect to lung cancer or pancreatic cancer, specifically in combination with other drugs). In embodiments, the presently disclosed fenoterol analogues may be administered with a combination of CTLA-4 blockade with PD-1 or PD-L1 inhibitors. In yet other embodiments, the presently disclosed fenoterol analogues may be administered with a therapeutic agent that targets an intrinsic checkpoint blockade, such as, for example, the gene encoding Cytokine-inducible SH2-containing protein (CISH).
[0097] In other examples, a disclosed pharmaceutical composition is administered as adjuvant therapy. For example, a pharmaceutical composition containing one or more of the disclosed compounds is administered orally daily to a subject in order to prevent or retard tumor growth. In one particular example, a composition containing equal portions of two or more disclosed compounds is provided to a subject. In one example, a composition containing unequal portions of two or more disclosed compounds is provided to the subject. For example, a composition contains unequal portions of a (R,R')-fenoterol derivative and a (S,R')-fenoterol derivative and/or a (R,S') -derivative. In one particular example, the composition includes a greater amount of the (S,R)- or (R,S')-fenoterol derivative. Such therapy can be given to a subject for an indefinite period of time to inhibit, prevent, or reduce tumor reoccurrence.
Methods of Use
[0098] The present disclosure includes methods of treating disorders including reducing or inhibiting one or more signs or symptoms associated with cancer, such as pancreatic cancer or breast cancer. Presently disclosed methods include administering fenoterol, such as (R,R)- fenoterol, a disclosed fenoterol analogue or a combination thereof (and, optionally, one or more other pharmaceutical agents) depending upon the receptor population of the tumor, to a subject in a pharmaceutically acceptable carrier and in an amount effective to reduce tumor generated L- lactate in the environment surrounding cancer cells. Treatment of a tumor includes preventing or reducing signs or symptoms associated with the presence of such tumor (for example, by reducing the size or volume of the tumor or a metastasis thereof). Such reduced growth can in some examples decrease or slow metastasis of the tumor, or reduce the size or volume of the tumor by at least 10%, at least 20%, at least 50%, or at least 75%, such as between 10%-90%, 20%-80%, 30%- 70%, 40%-60%, including a 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 90%, or 95% reduction. In another example, treatment includes reducing the invasive activity of the tumor in the subject, for example by reducing the ability of the tumor to metastasize. In some examples, treatment using the methods disclosed herein prolongs the time of survival of the subject.
[0099] Routes of administration useful in the disclosed methods include but are not limited to oral and parenteral routes, such as intravenous (IV), intraperitoneal (IP), rectal, topical, ophthalmic, nasal, and transdermal as described in detail above.
[00100] An effective amount of a disclosed fenoterol analogue will depend, at least, on the particular method of use, the subject being treated, the severity of the tumor, and the manner of administration of the therapeutic composition. A "therapeutically effective amount" of a composition is a quantity of a specified compound sufficient to achieve a desired effect in a subject being treated. For example, this may be the amount of a fenoterol analogue necessary to prevent or inhibit tumor growth and/or one or more symptoms associated with the tumor in a subject. Ideally, a therapeutically effective amount of a disclosed fenoterol analogue is an amount sufficient to prevent or inhibit a tumor, such as a brain or liver tumor growth and/or one or more symptoms associated with the tumor in a subject without causing a substantial cytotoxic effect on host cells.
[00101] Therapeutically effective doses of a disclosed fenoterol compound or pharmaceutical composition can be determined by one of skill in the art, with a goal of achieving concentrations that are at least as high as the IC50 of the applicable compound disclosed in the examples herein. An example of a dosage range is from about 0.001 to about 10 mg/kg body weight orally in single or divided doses. In particular examples, a dosage range is from about 0.005 to about 5 mg/kg body weight orally in single or divided doses (assuming an average body weight of approximately 70 kg; values adjusted accordingly for persons weighing more or less than average). For oral administration, the compositions are, for example, provided in the form of a tablet containing from about 1.0 to about 50 mg of the active ingredient, particularly about 2.5 mg, about 5 mg, about 10 mg, or about 50 mg of the active ingredient for the symptomatic adjustment of the dosage to the subject being treated. In one exemplary oral dosage regimen, a tablet containing from about 1 mg to about 50 mg active ingredient is administered two to four times a day, such as two times, three times or four times.
[00102] In other examples, a suitable dose for parental administration is about 1 milligram per kilogram (mg/kg) to about 100 mg/kg, such as a dose of about 10 mg/kg to about 80 mg/kg, such including about 1 mg/kg, about 2 mg/kg, about 5 mg/kg, about 20 mg/kg, about 30 mg/kg, about 40 mg/kg, about 50 mg/kg, about 80 mg/kg or about 100 mg/kg administered parenterally. However, other higher or lower dosages also could be used, such as from about 0.001 mg/kg to about 1 g/kg, such as about 0.1 to about 500 mg/kg, including about 0.5 mg/kg to about 200 mg/kg.
[00103] Single or multiple administrations of the composition comprising one or more of the disclosed compositions can be carried out with dose levels and pattern being selected by the treating physician. Generally, multiple doses are administered. In a particular example, the composition is administered parenterally once per day. However, the composition can be administered twice per day, three times per day, four times per day, six times per day, every other day, twice a week, weekly, or monthly. Treatment will typically continue for at least a month, more often for two or three months, sometimes for six months or a year, and may even continue indefinitely, i.e., chronically. Repeat courses of treatment are also possible. [00104] The specific dose level and frequency of dosage for any particular subject may be varied and will depend upon a variety of factors, including the activity of the specific compound, the metabolic stability and length of action of that compound, the age, body weight, general health, sex and diet of the subject, mode and time of administration, rate of excretion, drug combination, and severity of the condition of the subject undergoing therapy.
Selecting a Subject
[00105] Subjects can be screened prior to initiating the disclosed therapies, for example to select a subject in need of or at risk of developing cancer. Briefly, the method can include screening subjects to determine if they have or are at risk of developing cancer, such as if the subject is in need of pancreatic cancer or breast cancer inhibition. In embodiments, the cancer is regulated by at least one of B2-adrenergic receptor (AR) activity or expression, cannabinoid (CB) receptor activity or expression, or epidermal growth factor receptor (EGFR) activity or expression. Such cancers include, but are not limited to various types of breast cancer. Subjects having a tumor that expresses B2-adrenergic receptor (AR), cannabinoid (CB) receptor (including but not limited to GPR55), and epidermal growth factor receptor (EGFR) or at risk of developing such a tumor are selected. In one example, subjects are diagnosed with the tumor by clinical signs, laboratory tests, or both.
[00106] In exemplary embodiments, a subject in need of the disclosed therapies is selected by detecting a tumor expressing B2-adrenergic receptor (AR), cannabinoid (CB) receptor (including but not limited to GPR55), and epidermal growth factor receptor (EGFR) or regulated by their activity, such as by detecting B2-adrenergic receptor (AR) activity, cannabinoid (CB) receptor (including but not limited to GPR55) activity, and epidermal growth factor receptor (EGFR) activity or expression in a sample obtained from a subject identified as having, suspected of having or at risk of acquiring such a tumor. For example, detection of altered, such as at least a 10% alteration, including a 10%-90%, 20%-80%, 30%-70%, 40%-60%, such as a 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 90%, 95% alteration or more in B2-adrenergic receptor (AR) expression or activity, cannabinoid (CB) receptor (including but not limited to GPR55) expression or activity, and epidermal growth factor receptor (EGFR) expression or activity as compared to B2-adrenergic receptor (AR) expression or activity, cannabinoid (CB) receptor (including but not limited to GPR55) expression or activity, and epidermal growth factor receptor (EGFR) expression or activity in the absence of a primary tumor, indicates that the tumor can be treated using the fenoterol compositions and methods provided herein.
[00107] Pre-screening is not required prior to administration of the therapeutic agents disclosed herein (such as those including fenoterol, a fenoterol analogue or a combination thereof).
Assessment
[00108] Following the administration of one or more therapies, subjects can be monitored for decreases in tumor growth, tumor volume or in one or more clinical symptoms associated with the tumor. In particular examples, subjects are analyzed one or more times, starting 7 days following treatment. Subjects can be monitored using any method known in the art including those described herein including imaging analysis. Additional Treatments and Additional Therapeutic Agents
[00109] In particular examples, if subjects are stable or have a minor, mixed or partial response to treatment, they can be re-treated after re-evaluation with the same schedule and preparation of agents that they previously received for the desired amount of time, including the duration of a subject's lifetime. A partial response is a reduction, such as at least a 10%, at least a 20%), at least a 30%>, at least a 40%, at least a 50%, or at least a 70% reduction in one or more signs or symptoms associated with the disorder or disease, or activity, including tumor size or volume.
[00110] In some examples, the method further includes administering a therapeutic effective amount of a fenoterol analogue with additional therapeutic treatments. In particular examples, prior to, during, or following administration of a therapeutic amount of an agent that reduces tumor generated L-lactate in the environment surrounding the tumor, the subject can receive one or more other therapies. In one example, the subject receives one or more treatments to remove or reduce the tumor prior to administration of a therapeutic amount of a composition including fenoterol, a fenoterol analogue or combination thereof.
[00111] Examples of such therapies include, but are not limited to, surgical treatment for removal or reduction of the tumor (such as surgical resection, cryotherapy, or chemoembolization), as well as anti-tumor pharmaceutical treatments which can include radiotherapeutic agents, anti-neoplastic chemotherapeutic agents, antibiotics, alkylating agents and antioxidants, kinase inhibitors, and other agents. Particular examples of additional therapeutic agents that can be used include microtubule-binding agents, DNA intercalators or cross-linkers, DNA synthesis inhibitors, DNA and/or RNA transcription inhibitors, antibodies, enzymes, enzyme inhibitors, and gene regulators. These agents (which are administered at a therapeutically effective amount) and treatments can be used alone or in combination. Methods and therapeutic dosages of such agents are known to those skilled in the art, and can be determined by a skilled clinician.
[00112] "Microtubule-binding agent" refers to an agent that interacts with tubulin to stabilize or destabilize microtubule formation thereby inhibiting cell division. Examples of microtubule- binding agents that can be used in conjunction with the disclosed therapy include, without limitation, paclitaxel, docetaxel, vinblastine, vindesine, vinorelbine (navelbine), the epothilones, colchicine, dolastatin 15, nocodazole, podophyllotoxin and rhizoxin. Analogs and derivatives of such compounds also can be used and are known to those of ordinary skill in the art. For example, suitable epothilones and epothilone analogs are described in International Publication No. WO 2004/018478. Taxoids, such as paclitaxel and docetaxel, as well as the analogs of paclitaxel taught by U.S. Patent Nos. 6,610,860; 5,530,020; and/or 5,912,264 can be used.
[00113] The following classes of compounds may be of use in the methods described herein: DNA and/or RNA transcription regulators, including, without limitation, actinomycin D, daunorubicin, doxorubicin and derivatives and analogs thereof also are suitable for use in combination with the disclosed therapies; DNA intercalators and cross-linking agents that can be administered to a subject include, without limitation, cisplatin, carboplatin, oxaliplatin, mitomycins, such as mitomycin C, bleomycin, chlorambucil, cyclophosphamide and derivatives and analogs thereof; DNA synthesis inhibitors suitable for use as therapeutic agents include, without limitation, methotrexate, 5-fluoro-5'-deoxyuridine, 5-fluorouracil and analogs thereof. (Examples of suitable enzyme inhibitors include, without limitation, camptothecin, etoposide, formestane, trichostatin and derivatives and analogs thereof. Examples of alkylating agents include carmustine or lomustine.); compounds that affect gene regulation include agents that result in increased or decreased expression of one or more genes, such as raloxifene, 5- azacytidine, 5-aza-2'-deoxycytidine, tamoxifen, 4-hydroxytamoxifen, mifepristone and derivatives and analogs thereof; and kinase inhibitors include Gleevac, Iressa, and Tarceva that prevent phosphorylation and activation of growth factors.
[00114] Other therapeutic agents, for example anti -tumor agents, that may or may not fall under one or more of the classifications above, also are suitable for administration in combination with the disclosed therapies. By way of example, such agents include adriamycin, apigenin, rapamycin, zebularine, cimetidine, and derivatives and analogues thereof.
[00115] In one example, at least a portion of the tumor is surgically removed (for example via cryotherapy), irradiated, chemically treated (for example via chemoembolization) or combinations thereof, prior to administration of the disclosed therapies (such as administration of fenoterol, a fenoterol analogue or a combination thereof). For example, a subject can have at least a portion of the tumor surgically excised prior to administration of the disclosed therapies. In an example, one or more chemotherapeutic agents are administered following treatment with a composition including fenoterol, a fenoterol analogue or a combination thereof.
[00116] The subject matter of the present disclosure is further illustrated by the following non- limiting Examples.
[00117] Materials and Methods
[00118] The material and methods used for the following Examples were as follows: [00119] Materials. (R,R ')-, (R,S)' -, (S,R )- and (S,S')-fenoterol and the fenoterol analogs, (R,R')-ethylfenoterol, (R,R')-4'-aminofenoterol, (R,R')-l-naphthylfenoterol and (R,R)- and (R,S')-4'-methoxy-l-naphthylfenoterol, were synthesized as previously described (Jozwiak et al, J Med Chem 50:2903-2915, 2007; Jozwiak et al, Bioorg Med Chem 18:728-736, 2010; each of which is incorporated by reference in its entirety). [3H]-Thymidine (70-90 Ci/mmol) was purchased from PerkinElmer Life and Analytical Sciences (Waltham, MA). Eagle's Minimum Essential Medium (E-MEM), trypsin solution, phosphate-buffered saline (PBS), fetal bovine serum (FBS), 100X solutions of sodium pyruvate (100 mM), L-glutamine (200 mM), and penicillin/streptomycin (a mixture of 10,000 units/ml penicillin and 10,000 μg/ml streptomycin) were obtained from Quality Biological (Gaithersburg, MD). WIN 55,212-2, AM251, and AM630 were purchased from Cayman Chemical (Ann Arbor, MI). ICI 118,551 hydrochloride and (R)-isoproterenol were obtained from Sigma-Aldrich (St. Louis, MO). Phenylmethylsulfonyl fluoride (PMSF), benzamidine, leupeptin, pepstatin A, MgCl2, EDTA, Trizma-Hydrochloride (Tris-HCl), (±) -propranolol and minimal essential medium (MEM) were obtained from Sigma Aldrich (St. Louis, MO). Egg phosphatidylcholine lipids (PC) were obtained from Avanti Polar Lipids (Alabaster, AL). (±)-fenoterol was purchased from Sigma - Aldrich and [3H]-(±)-fenoterol was acquired from Amersham Biosciences (Boston, MA). The organic solvents n-hexane, 2-propanol and triethylamine were obtained as ultra pure HPLC grade solvents from Carlo Erba (Milan, Italy). Fetal bovine serum and penicillin-streptomycin were purchased from Life Technologies (Gaithersburg, MD), and [125I]- (i)-iodocyanopindolol (ICYP) was purchased from NEN Life Science Products, Inc. (Boston, MA). Methods
[00120] Effect of (R.S VMNF on Cancer Biomarker Expression in PANC-1 cells.
PANC-1 cells were incubated with media containing vehicle (0.01% DMSO) or (R,S')-MNF (0- 10 μΜ) for 24h. The medium was removed, and cells were collected and processed for immunoblot analysis as recently described (Singh et al., Pharmacological Research 111, pages 757-766, 2016). All membrane-bound primary antibodies were detected with horseradish peroxidase-conjugated secondary antibodies (Santa Cruz Biotechnology, Dallas, TX) and visualized by enhanced chemiluminescence (ECL Plus, GE Healthcare, Piscataway, NT). Quantification of the protein bands was performed by volume densitometry using ImageJ software (National Institutes of Health, Bethesda, MD) and normalization to β-actin or Lamin A/C. Primary antibodies used in this study were raised against EGFR (sc-03) and β-catenin (sc- 7199) (Santa Cruz Biotechnology); PKM2 (ab38237) and β-actin (ab6276) (Abeam, Cambridge, MA). Three independent experiments were conducted on three separate days.
[00121] PANC-1 tumor xenografts in mice. Female Balb/c nude mice (aged between 6-8 weeks, weight 18-20 g) were purchased from HFK Bioscience Co., Ltd. (Beijing, China) and maintained under pathogen-free conditions with a 12-hour light/12-hour dark cycle. Animals had free access to drinking water and were fed ad libitum with normal chow. Each mouse was inoculated subcutaneously at the right flank region with PANC-1 cells (5 x 106) in O. lmL of PBS for tumor development. All animals were weighed and the tumor volumes measured in two dimensions using a caliper, and the volume expressed in mm3 using the formula: V = 0.5 a x b2, where a and b are the long and short diameters of the tumor, respectively. The treatments were started at Day 8 when the mean tumor size reached 139 ± 34 mm3 and the mice weighed 21.1 ± 1.1 g. The mice were assigned into groups (n = 10) using randomized block design based on their tumor volumes. The mice received a single intraperitoneal (ip) injection 5 days per week for 3 treatment cycles of either vehicle (1% hydroxypropyl-P-cyclodextrin) (Control), 20mg-kg_1 (R,S')-MNF (Arm 2) or 40mg-kg_1 (R,S')-MNF (Arm 2). The dosing volume was adjusted according to weight (lC^L/g). Tumor volumes and weights were determined at the beginning and end of each dosing cycle, while visual estimation of food and water consumption, eye/hair matting, and behavior such as mobility were determined daily. The animals were euthanized by cervical extension 5 days after the end of the last dosing cycle. Plasma samples were collected at the conclusion of the studies and, after collection of plasma samples, the tumors were excised, weighed, divided into three portions and snap frozen. The xenograft studies were conducted at the Crown Biosciences, Ltd. facilities and all protocols were approved by the Animal Care and Use Committee at CrownBio (AN- 1407-009- 164), which are based on "the Guide for the Care and Use of Laboratory Animals" (NRC 2011).
[00122] Determination of (R,S')-MNF concentrations in plasma and tumor tissues. The concentrations of (R,S')-MNF in plasma and tumor samples obtained from Arm 2 and Arm 3 were analyzed by LC-MS as described below.
[00123] Determination of L-lactate plasma concentrations. The concentration of L-lactate in plasma samples obtained from Control 1, Arm 1 and Arm 2 of the study were measured using the Lactate Colorimetric Assay Kit II (Sigma-Aldrich, St. Louis, MO). The determinations were carried out according to the manufacturer's instructions.
[00124] Statistical analysis. Sigmoidal dose-response curves (IC50 curves) were determined using the 'nonlinear regression (curve fit)' model contained within the Prism 4 software package (GraphPad Software, Inc) running on a personal computer. For immunoblot analyses, tumor volume, and animal weight analyses, statistical comparisons between treated and control groups were performed using unpaired Student's t-tests. P values < 0.05 were considered significant.
[00125] Experimental Methods used in studies with (RiO-MNF. PANC-1 Tumor
Xenograft in Mice. Female Balb/c nude mice (aged between 6-8 weeks, weight 18-20 g) were purchased from HFK Bioscience Co., Ltd. (Beijing, China) and maintained under pathogen-free conditions with a 12-h light/12-h dark cycle. Animals had free access to drinking water and were fed ad libitum with normal chow. Each mouse was inoculated subcutaneously at the right flank region with PANC-1 cells (5- 106) in O. lmL of PBS for tumor development. All animals were weighed and the tumor volumes measured in two dimensions using a caliper, and the volume expressed in mm3 using the formula: V = 0.5 a x b2, where a and b are the long and short diameters of the tumor, respectively. The treatments were started at day 8 when the mean tumor size reached 164 mm3, at which time the mice were assigned into two groups (n = 10) using randomized block design based on their tumor volumes. Mice received an i.p. injection of either vehicle (20% hydroxypropyl-P-cyclodextrin) (Control) or 10 mg-kg"1 MNF once daily for 16 days adjusted to 25 mg-kg"1 for the last 5 days (Experimental). The dosing volume was adjusted according to weight (10 μΕ-g-l). At the end of the study, the animals were euthanized by cervical extension and the tumors were collected, weighed, divided into three portions and snap frozen. All protocols were approved by the Animal Care and Use Committee at CrownBio (AN- 1407-009-164), which are based on "the Guide for the Care and Use of Laboratory Animals" (NRC 2011). [00126] Western Blot Analysis. Cells and frozen tumor tissues were lysed in radioimmunoprecipitation buffer containing EGTA and EDTA (Boston BioProducts, Ashland, MA, USA) supplemented with protease inhibitor cocktail (Sigma-Aldrich). Protein concentration in clarified lysates was determined using the bicinchoninic acid reagent (Thermo Fisher Scientific, Waltham, MA, USA). Proteins (20 μg/well) were separated on 4-12% precast gels (Invitrogen, Carlsbad, CA, USA) using SDS-polyacrylamide gel electrophoresis under reducing conditions and then electrophoretically transferred onto polyvinylidene fluoride membrane (Invitrogen). Western blots were performed according to standard methods, which involved a blocking step in Tris-buffered saline/0.1% Tween-20 (TBS-T) supplemented with 5% non-fat milk and incubation with primary antibodies of interest. All antibodies were detected with horseradish peroxidase-conjugated secondary antibodies (Santa Cruz Biotechnology, Dallas, TX, USA) and visualized by enhanced chemiluminescence (ECL Plus, GE Healthcare, Piscataway, NJ, USA). Quantification of the protein bands was performed by volume densitometry using ImageJ software (National Institutes of Health, Bethesda, MD, USA) and normalization to β- actin. The primary antibodies used in this study were raised against EGFR (sc-03), MCT4 (sc- 50329), Glut8 (sc-30108), and β-catenin (sc-7199) (Santa Cruz Biotechnology); PKM2 (ab38237) and β-actin (ab6276) (Abeam, Cambridge, MA, USA); hexokinase II (cat. #2867) and PDKl (cat. #3062S) (Cell Signaling Technology, Beverly, MA, USA). The antibodies were used at the dilution recommended by the manufacturers.
[00127] Liquid Chromatography-Mass Spectrometry (LC/MS) Analysis. For the determination of intracellular lactate, 3-hydroxbutyrate and carnitine, PANC-1 cells were collected by scrapping in PBS and centrifuged at 1000 rpm for 5 min. Cell pellets were suspended in 20 μΙ_, water followed by the addition of 10 μΙ_, of 100 μΜ p-aminohippuric acid as an internal standard. The resulting mixture was vortexed for 1 min and then 80 μΙ_, of methanol was added prior to sonication (PRO200 homogenizer, PRO Scientific, Oxford, CT, USA) for 10 min on ice. The mixture was centrifuged for 15 min at 14000 rpm at 4 °C and the supernatant collected and analyzed using a system composed of an Agilent Technologies 1100 LC/MSD equipped with a G1322A degasser, G1312A quaternary pump, G1367A autosampler, G1316A column thermostat and G1946D mass spectrometer supplied with electrospray ionization (ESI). Selected ion monitoring (SIM) chromatograms were acquired using ChemStation software. For the separation of compounds, a reverse phase Zorbax SB C18 column (150 x 2.1 mm, Agilent Technologies, Palo Alto, CA, USA) was used and operated at 25 °C. Gradient elution was used for the separation, with solvent (A) composed of 0.1 % formic acid in water and solvent (B) 0.1 % formic acid in acetonitrile. The solvent gradient in volumetric ratios of solvents A and B was as followed: 0-2 min, 100 A/0 B; 2-20 min, 20 A/80 B; 20-27 min, 20 A/80 B; 27-38 min, 100 A/0 B; 38-50 min, 100 A/0 B. The flow rate was 0.8 mL-min"1 and the injection volume was 20 μΐ,. The compounds of interest were monitored in the positive-ion mode for SIM at m/z 162.1 (carnitine), m/z 90.1 (lactate) and m/z 105.1 (3-hydroxybutyrate). The internal standard was monitored at m/z 195.1 (p-aminohippuric acid).
[00128] The C6 cell line is derived from a rat glioblastoma brain tumor and is a standard model for brain tumors. In accordance with the methods described herein (R,R')-M F decreases glucose uptake and L-lactate output in this cell line, expanding the effect to more than one type of tumor. Experimental
[00129] Cell Culture. The human pancreatic tumor cell line, PANC-1, breast cancer cell lines MDA-MB-231 and MCF-7, and rat-derived C6 glioblastoma cell line were purchased from ATCC (Manassas, VA, USA). Upon receipt of the three cell lines, cells were expanded for a few passages to enable the generation of new frozen stocks. Cells were resuscitated as needed and used for fewer than 6 months after resuscitation (no more than 10 passages). ATCC performs thorough cell line authentication utilizing Short Tandem Repeat (STR) profiling.
[00130] PANC-1 and C6 cells were maintained in DMEM with L-glutamine supplemented with 10% FBS and 1% penicillin/streptomycin. MDA-MB-231 cells were maintained in RPMI- 1640 supplemented with 10% FBS and 1% penicillin/streptomycin and MCF-7 cells were maintained in EMEM with L-glutamine supplemented with 10% FBS and 0.01 mg-ml"1 human recombinant insulin. Cells were maintained in a controlled environment (37 °C under humidified 5% C02 in air), and the medium was replaced every 2-3 days. Prior to experiments, cells were seeded on 100 x 20 mm tissue culture plates and grown to -70% confluency unless stated otherwise.
[00131] Cell Treatment. In the first series of experiments, the original media was replaced with media containing vehicle (0.01% DMSO) or MNF (0.01, 0.10, 0.50 and 1.00 μΜ) for 24 h. The medium was removed, and cells were collected and processed for immunoblot analysis. In a second series of experiments, PANC-1 cells were treated with vehicle (0.01% DMSO) or MNF (0.50 and 1.00 μΜ) for 24 h after which cells were washed, and then collected and processed for NMR or LC/MS analysis. In the last series of experiments, MDA-MB-231 and MCF-7 cells were treated with vehicle (0.01% DMSO) or MNF (1.00 μΜ) for 24 h and processed for LC/MS analysis. All experiments were repeated three times on three separate days, unless stated otherwise.
[00132] Glucose consumption and lactate production in C6 cells. To assess glucose consumption and lactate production, C6 cells were culture in 48-well plates and treated with MNF (0.02 or 0.20 μΜ) or vehicle (DMSO, 0.1%) and glucose and lactate concentrations in the incubation media were periodically monitored for 48 h. At the end of each incubation, the culture media were collected to determine glucose and lactate content using the Liquick Cor-Glucose and Liquick Cor-Lactate Diagnostic Kits (Cormay, Lublin, Poland). Measurements were carried out according to the manufacturer's protocols.
Results
[00133] Functional effects of MNF on glucose uptake and L-lactate output in C6 cells. C6 cells were incubated with MNF (0.02 and 0.2 mM) and aliquots of the spent medium were collected at various time points over a 48-h period for the measure of glucose and L-lactate. Glucose uptake and L-lactate output are two classical readouts of the glycolytic pathway. Treatment with MNF elicited an accumulation of glucose in the spent media of C6 together with reduction in L-lactate levels (Figure 3) compared with the spent media from untreated cells (P < 0.001 for both metabolites). The same profile of glucose consumption and L-lactate output was obtained regardless of the MNF dose.
[00134] There was no significant difference in pre-dose L-lactate concentrations between control animals and experimental cohorts and there was no significant change in L-lactate concentration from pre-dose to termination in control animals. [00135] While several embodiments of the disclosure have been described, it is not intended that the disclosure be limited thereto, as it is intended that the disclosure be as broad in scope as the art will allow and that the specification be read likewise. Other elements, steps, methods and techniques that are insubstantially different from those described above and/or in the appended claims are also intended to be within the scope of the disclosure. Therefore, the above description should not be construed as limiting, but merely as exemplifications of presently disclosed embodiments. Thus the scope of the embodiments should be determined by the appended claims and their legal equivalents, rather than by the examples given.
[00136] Persons skilled in the art will understand that the materials and methods specifically described herein and illustrated in the accompanying drawings are non-limiting exemplary embodiments. The features illustrated or described in connection with one exemplary embodiment may be combined with the features of other embodiments. Such modifications and variations are intended to be included within the scope of the present disclosure. As well, one skilled in the art will appreciate further features and advantages of the present disclosure based on the above-described embodiments. Accordingly, the present disclosure is not to be limited by what has been particularly shown and described, except as indicated by the appended claims.

Claims

WHAT IS CLAIMED IS:
1. A method comprising:
increasing a patient's response to an immune checkpoint blockade therapy by administering a therapeutically effective amount of a pharmaceutical composition containing a pharmaceutically acceptable carrier and a fenoterol analogue to a subject to treat cancer.
2. A method comprising:
reducing tumor generated L-lactate in a microenvironment surrounding a tumor by administering a pharmaceutical composition containing a pharmaceutically acceptable carrier and a fenoterol analogue to a subject to treat cancer.
3. The method of claim 2 wherein the fenoterol analogue is a PKM2 antagonist.
4. The method of claim 2 wherein the fenoterol analogue decreases HK2 expression and activity.
5. The method of claim 2 wherein the fenoterol analogue attenuates monocarboxylate transporter 4 (MCT4) expression and/or function.
6. The method of claim 2 wherein the pharmaceutical composition reduces tumor generated L-lactate in the microenvironment of either a breast cancer tumor or a pancreatic cancer tumor.
7. The method of claim 2 further comprising administering an immune checkpoint blockade therapy before, during or after administration of the pharmaceutical composition.
8. A method comprising:
decreasing L-lactate export out of cancer cells by administering a pharmaceutical composition containing a pharmaceutically acceptable carrier and a fenoterol analogue to a subject to treat cancer.
9. The method of claim 8 wherein the fenoterol analogue attenuates monocarboxylate transporter 4 (MCT4) expression and/or function.
10. A method comprising:
increasing L-lactate concentrations in cancer cells by administering a pharmaceutical composition containing a pharmaceutically acceptable carrier and a fenoterol analogue to a subject to treat cancer.
11. The method of claim 10 wherein the fenoterol analogue attenuates monocarboxylate transporter 4 (MCT4) expression and/or function.
12. A method comprising:
restoring oxidative glucose metabolism in a cancer cell by administering a pharmaceutical composition containing a pharmaceutically acceptable carrier and a fenoterol analogue to a subject to treat cancer.
13. The method of claim 12 wherein the fenoterol analogue inhibits expression of hexokinase-2 (HK2) in cancer cells showing increased HK2 expression.
14. A method comprising:
increasing the sensitivity of cancer cells to a cell death inducer by administering a pharmaceutical composition containing a pharmaceutically acceptable carrier and a fenoterol analogue to a subject to treat cancer.
15. The method of claim 14 wherein the fenoterol analogue inhibits expression of hexokinase-2 (HK2) in cancer cells showing increased HK2 expression.
16. A method comprising:
decreasing expression of one or more glucose transporter facilitator in cancer cells by administering a pharmaceutical composition containing a pharmaceutically acceptable carrier and a fenoterol analogue to a subject to treat cancer.
17. The method of claim 16 wherein the pharmaceutical composition decreases expression of Glut-1.
18. The method of claim 16 wherein the pharmaceutical composition decreases expression of Glut-8.
19. The method of any of the preceding claims wherein the pharmaceutical composition administered contains one or more compounds selected from the group consisting of (R,R')-4'- methoxy-l-naphthylfenoterol ("MNF"), (R,S')-4'-methoxy-l-naphthylfenoterol, (R,R')- ethylMNF, (R,R')-napthylfenoterol, (R,S')-napthylfenoterol, (R,R')-ethyl-naphthylfenoterol, (R,R' )-4 ' -amino- 1 -naphthylfenoterol, (R,R' )-4 ' -hydroxy- 1 -naphthylfenoterol, (R,R' )-4 ' - methoxy-ethylfenoterol, (R,R')-4'-methoxyfenoterol, (R,R')-ethylfenoterol, (R,R')-fenoterol and their respective stereoisomers.
20. The method of any of the preceding claims wherein the pharmaceutical composition administered contains a compound of the formula:
21. The method of any of the preceding claims further comprising administering an additional therapeutic treatment before, during or after administration of the pharmaceutical composition.
22. The method of any of the preceding claims further comprising administering an additional agent for the treatment of cancer before, during or after administration of the pharmaceutical composition.
PCT/US2018/015874 2017-01-30 2018-01-30 Methods of treating cancer WO2018140923A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201762451847P 2017-01-30 2017-01-30
US62/451,847 2017-01-30

Publications (1)

Publication Number Publication Date
WO2018140923A1 true WO2018140923A1 (en) 2018-08-02

Family

ID=62978734

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2018/015874 WO2018140923A1 (en) 2017-01-30 2018-01-30 Methods of treating cancer

Country Status (1)

Country Link
WO (1) WO2018140923A1 (en)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016144371A1 (en) * 2015-03-06 2016-09-15 Mitchell Woods Pharmaceuticals, Inc. Methods of treating cancer
WO2016145427A1 (en) * 2015-03-12 2016-09-15 Health Research, Inc. COMBINATION OF β-ADRENERGIC RECEPTOR ANTAGONISTS AND CHECK POINT INHIBITORS FOR IMPROVED EFFICACY AGAINST CANCER

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016144371A1 (en) * 2015-03-06 2016-09-15 Mitchell Woods Pharmaceuticals, Inc. Methods of treating cancer
WO2016145427A1 (en) * 2015-03-12 2016-09-15 Health Research, Inc. COMBINATION OF β-ADRENERGIC RECEPTOR ANTAGONISTS AND CHECK POINT INHIBITORS FOR IMPROVED EFFICACY AGAINST CANCER

Non-Patent Citations (8)

* Cited by examiner, † Cited by third party
Title
CALVO, MB ET AL.: "Potential Role of Sugar Transporters in Cancer and Their Relationship with Anticancer Therapy", INTERNATIONAL JOURNAL OF ENDOCRINOLOGY, vol. 2010, 2010, pages 1 - 15, XP055060456 *
LABAK, CM ET AL.: "Glucose transport: meeting the metabolic demands of cancer, and applications in glioblastoma treatment", AMERICAN JOURNAL OF CANCER RESEARCH, vol. 6, no. 8, 2016, pages 1599 - 1608, XP055531817 *
MENG, X ET AL.: "The combination of Paclitaxel and Gefitinib inhibits endometrial cancer cells by inducing mitotic catastrophe: proof of principle for dual therapy in endometrial cancer", PROCEEDINGS IN OBSTETRICS AND GYNECOLOGY, vol. 2, no. 1, 2011, pages 1 - 2, XP055521127 *
MOMCILOVIC, M ET AL.: "Targeted inhibition of EGFR and glutaminase induces metabolic crisis in EGFR mutant lung cancer", CELL REPORTS, vol. 18, no. 3, 17 January 2017 (2017-01-17), pages 1 - 18, XP055519204 *
SINGH, NS ET AL.: "Abstract B32: GRP55 antagonists alter tumor microenvironment and inhibit tumor growth in a pancreatic tumor xenograft model", CANCER RESEARCH, vol. 76, no. 24, XP055521096 *
WAGNER, M ET AL.: "Expression of a Truncated EGF Receptor Is Associated with Inhibition of Pancreatic Cancer Cell Growth and Enhanced Sensitivity to Cisplatinum", INTERNATIONAL JOURNAL OF CANCER, vol. 68, 1996, pages 782 - 787, XP055531816 *
WOLF, A ET AL.: "Hexokinase 2 is a key mediator of aerobic glycolysis and promotes tumor growth in human glioblastoma multiforme", THE JOURNAL OF EXPERIMENTAL MEDICINE, vol. 208, no. 2, 2011, pages 313 - 326, XP055521122 *
ZHANG, H ET AL.: "Lectin PCL inhibits the Warburg effect of PC3 cells by combining with EGFR and inhibiting HK 2", ONCOLOGY REPORTS, vol. 37, no. 3, 16 January 2017 (2017-01-16), pages 1765 - 1771, XP055521100 *

Similar Documents

Publication Publication Date Title
US10617654B2 (en) Use of fenoterol and fenoterol analogues in the treatment of glioblastomas and astrocytomas
US10772849B2 (en) Methods of regulating cannabinoid receptor activity-related disorders and diseases
US20180042867A1 (en) Methods of treating cancer
WO2017059268A1 (en) Methods of reducing chemoresistance and treating cancer
US20190076379A1 (en) Methods of treating breast cancer
WO2018140923A1 (en) Methods of treating cancer
US20170172944A1 (en) Methods of treating melanoma
US8729053B2 (en) Nuclear factor kappa B pathway inhibitor composition and use of same
AU2016341109A1 (en) Compositions and methods of regulating cancer related disorders and diseases
WO2012131096A1 (en) Novel heterocyclic compounds as modulators of sphingolipid signaling and uses thereof

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 18745187

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 18745187

Country of ref document: EP

Kind code of ref document: A1